US20090111160A1 - Compositions comprising bowman-birk protease inhibitors and variants thereof - Google Patents

Compositions comprising bowman-birk protease inhibitors and variants thereof Download PDF

Info

Publication number
US20090111160A1
US20090111160A1 US12/172,921 US17292108A US2009111160A1 US 20090111160 A1 US20090111160 A1 US 20090111160A1 US 17292108 A US17292108 A US 17292108A US 2009111160 A1 US2009111160 A1 US 2009111160A1
Authority
US
United States
Prior art keywords
seq
bbi
loop
sequence
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US12/172,921
Other versions
US7696173B2 (en
Inventor
Katherine Collier
Grant Ganshaw
Hans De Nobel
Scott D. Power
Anita Van Kimmenade
Marc Kolkman
Jeffrey Miller
Brian Schmidt
Gudrun Vogtentanz
David Estell
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Danisco US Inc
Original Assignee
Danisco US Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Danisco US Inc filed Critical Danisco US Inc
Priority to US12/172,921 priority Critical patent/US7696173B2/en
Assigned to GENENCOR INTERNATIONAL, INC. reassignment GENENCOR INTERNATIONAL, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POWER, SCOTT, ESTELL, DAVID, GANSHAW, GRANT, DE NOBEL, HANS, KOLKMAN, MARC, MILLER, JEFFREY, COLLIER, KATHERINE, SCHMIDT, BRIAN, VAN KIMMENADE, ANITA, VOGTENTANZ, GUDRUN
Publication of US20090111160A1 publication Critical patent/US20090111160A1/en
Assigned to DANISCO US INC. reassignment DANISCO US INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: GENENCOR INTERNATIONAL, INC.
Application granted granted Critical
Publication of US7696173B2 publication Critical patent/US7696173B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/811Serine protease (E.C. 3.4.21) inhibitors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species.
  • the present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • proteases are involved in a wide variety of biological processes. Disruption of the balance between proteases and protease inhibitors is often associated with pathologic tissue destruction. Indeed, various studies have focused on the role of proteases in tissue injury, and it is thought that the balance between proteases and protease inhibitors is a major determinant in maintaining tissue integrity. Serine proteases from inflammatory cells, including neutrophils, are implicated in various inflammatory disorders, such as pulmonary emphysema, arthritis, atopic dermatitis and psoriasis.
  • Proteases also appear to function in the spread of certain cancers. Normal cells exist in contact with a complex protein network, called the extracellular matrix (ECM).
  • ECM extracellular matrix
  • the ECM is a barrier to cell movement and cancer cells must devise ways to break their attachments, degrade, and move through the ECM in order to metastasize.
  • Proteases are enzymes that degrade other proteins and have long been thought to aid in freeing the tumor cells from their original location by chewing up the ECM. Recent studies have suggested that they may promote cell shape changes and motility through the activation of a protein in the tumor cell membrane called Protease-Activated Receptor-2 (PAR2). This leads to a cascade of intracellular reactions that activates the motility apparatus of the cell.
  • PAR2 Protease-Activated Receptor-2
  • one of the first steps in tumor metastasis is a reorganization of the cell shape, such that it forms a distinct protrusion at one edge facing the direction of migration.
  • the cell then migrates through a blood vessel wall and travels to distal locations, eventually reattaching and forming a metastatic tumor.
  • human prostatic epithelial cells constitutively secrete prostate-specific antigen (PSA), a kallikrein-like serine protease, which is a normal component of the seminal plasma.
  • PSA prostate-specific antigen
  • protease acts to degrade the extracellular matrix and facilitate invasion of cancerous cells.
  • protease inhibitors have been shown to inhibit tumor promotion in vivo and in vitro. Previous investigations have indicated that certain protease inhibitors belonging to a family of structurally-related proteins classified as serine protease inhibitors or SERPINS, are known to inhibit several proteases including trypsin, cathepsin G, thrombin, and tissue kallikrein, as well as neutrophil elastase.
  • SERPINS serine protease inhibitors
  • SERPINS serine protease inhibitors
  • Topical administration of protease inhibitors finds use in such conditions as atopic dermatitis, a common form of inflammation of the skin, which may be localized to a few patches or involve large portions of the body.
  • the depigmenting activity of protease inhibitors and their capability to prevent ultraviolet-induced pigmentation have been demonstrated both in vitro and in vivo (See e.g., Paine et al., J. Invest. Dermatol., 116: 587-595 [2001]).
  • Protease inhibitors have also been reported to facilitate wound healing. For example, secretory leukocyte protease inhibitor was demonstrated to reverse the tissue destruction and speed the wound healing process when topically applied.
  • serine protease inhibitors can also help to reduce pain in lupus erythematosus patients (See e.g., U.S. Pat. No. 6,537,968).
  • protease inhibitors interfere with the action of proteases.
  • Naturally occurring protease inhibitors can be found in a variety of foods such as cereal grains (oats, barley, and maize), Brussels sprouts, onion, beetroot, wheat, finger millet, and peanuts.
  • One source of interest is the soybean.
  • the average level of protease inhibitors present in soybeans is around 1.4 percent and 0.6 percent for Kunitz and Bowman-Birk respectively, two of the most important protease inhibitors. Notably, these low levels make it impractical to isolate the natural protease inhibitor for clinical applications.
  • the present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species.
  • the present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • nucleic acids Provided herein are nucleic acids, cells and methods for the production of protease inhibitors and variants thereof.
  • the present invention provides nucleic acids encoding at least one functional protease inhibitor.
  • a nucleic acid comprising regulatory sequences operatively linked to a first, second, third and fourth nucleic acid sequences are provided.
  • terminator sequences are provided following the fourth nucleic acid sequence.
  • the first nucleic acid sequence encodes a signal polypeptide functional as a secretory sequence in a first host organism
  • the second nucleic acid encodes a secreted polypeptide or functional portion thereof normally secreted from the first or a second host organism
  • the third nucleic acid encodes a cleavable linker
  • the fourth nucleic acid encodes a protease inhibitor or fragment thereof.
  • the present invention provides at least one expression cassette comprising nucleic acid sequences encoding at least one protease inhibitor.
  • the present invention provides polynucleotides encoding at least one protease inhibitor variant.
  • the polynucleotide encodes a Bowman-Birk Inhibitor (BBI) variant, wherein at least one loop of the wild-type BBI has been altered.
  • BBI Bowman-Birk Inhibitor
  • the present invention also provides methods of expressing functional protease inhibitors or variants thereof.
  • host cells suitable for production of functional protease inhibitors and/or variants thereof are provided.
  • a host cell is (i) transformed with at least one expression cassette comprising a nucleic acid sequence encoding at least one protease inhibitor or variant thereof, and (ii) cultured under appropriate conditions to express at least one protease inhibitor or variants thereof.
  • the method further comprises recovering the protease inhibitor or variant thereof.
  • a host cell is (i) transformed with a first expression cassette comprising at least one nucleic acid sequence encoding a protease inhibitor or variant thereof, (ii) transformed with a second expression cassette comprising a nucleic acid sequence encoding at least one thiol-disulfide oxidoreductase or chaperone, and (iii) cultured under appropriate conditions to express the protease inhibitors or variant thereof.
  • the protease inhibitors or variants thereof are recovered.
  • the protease inhibitors or variant thereof are expressed as a fusion protein.
  • the methods further comprise recovering the protease inhibitor or variant thereof.
  • functional protease inhibitors and variants thereof are provided.
  • the functional protease inhibitor or variant thereof is expressed as a fusion protein consisting of the signal sequence, a cellulase catalytic domain, a cleavable linker region, and then by the mature protease inhibitor or variant thereof.
  • the expressed proteins are treated with a protease and/or acid/heat treatment to liberate a protease inhibitor or variant thereof from the fusion protein.
  • the present invention provides a polypeptide having protease inhibitory activity, selected from the group consisting of
  • a scaffold comprising at least one variant sequence.
  • compositions comprising a fusion protein, wherein the fusion protein comprises a protease inhibitor, and a peptide of interest.
  • the fusion protein comprises an amino acid selected from the group consisting of SEQ ID NOS:2, 4, 6 and 8.
  • the fusion protein is encoded by a nucleotide sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, and 7.
  • the protease inhibitor is selected from the group consisting of Bowman-Birk inhibitor (BBI), soybean trypsin inhibitor (STI), and eglin C.
  • the protease inhibitor is BBI, and wherein the BBI comprises at least one loop selected from the group consisting of a trypsin loop and a chymotrypsin loop.
  • the protease inhibitor is a scaffold for the peptide of interest.
  • the loop comprises at least one peptide of interest.
  • the peptide of interest comprises the amino acid sequence set forth in SEQ ID NO:9.
  • the fusion protein comprises the amino acid sequence set forth in SEQ ID NO:4.
  • the present invention also provides methods for producing at least one protease inhibitor in a bacterial cell comprising: a) introducing a DNA construct into a bacterial cell, wherein the DNA construct comprises a heterologous DNA sequence encoding a protease inhibitor derived from a Bowman-Birk Inhibitor (BBI) or variants thereof; b) culturing the bacterial cell under suitable culture conditions to allow expression of the heterologous DNA sequence; and c) producing the protease inhibitor.
  • the construct further comprises at least one catalytic domain.
  • catalytic domain is selected from the group consisting of cellulase, cutinase, and disulfide isomerase.
  • the method further comprises the step of recovering the protease inhibitor. In some particularly preferred embodiments, the method further comprises the step of activating the protease inhibitor. In some most preferred embodiments, the activating is accomplished by exposing the protease inhibitor to at least one reagent selected from the group consisting of compositions that reduce disulfide bonds, compositions that oxidize disulfide bonds, and compositions that alter the redox potential. In some embodiments, the bacterial cell is a member of the genus Bacillus .
  • protease inhibitor is selected from the group consisting of: i) a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 11; and ii) a protease inhibitor having at least 90% sequence identity with SEQ ID NO:13.
  • the protease inhibitor comprises a variant sequence.
  • the protease inhibitor is Bowman-Birk inhibitor and wherein SEQ ID NO:9 is substituted for at least one loop, wherein the loop is selected from the group consisting of the trypsin loop and the chymotrypsin loop.
  • the method further comprises the step of introducing a second nucleic acid sequence encoding a thiol-disulfide oxidoreductase or chaperone into the bacterial cell.
  • the protease inhibitor is expressed as a fusion protein.
  • the fusion protein further comprises a cellulase catalytic domain, a cleavage site, and the protease inhibitor.
  • the fusion protein is processed by a protease or acid/heat treatment to liberate the protease inhibitor.
  • the fusion protein further comprises at least one linker sequence.
  • the linker sequence is selected from the group consisting of SEQ ID NOS:141-143.
  • the present invention also provides protease inhibitor compositions comprising the protease inhibitor produced according to any and all of the methods described herein.
  • the various primers and oligonucleotides described herein find use in the production of the fusion proteins of the present invention.
  • the present invention provides numerous peptides suitable for use in the present invention.
  • the present invention also provides methods for inhibiting the proteolytic activity of a target protein comprising contacting the target protein with any of the protease inhibitor compositions described herein, and binding the target protein wherein the proteolytic activity of the target protein i inhibited.
  • the present invention further provides isolated polynucleotides encoding a protease inhibitor selected from the group consisting of polypeptide sequences set forth in SEQ ID NOS:10 and 12.
  • the present invention also provides expression vectors comprising a polynucleotide sequence, wherein the polynucleotide sequence is selected from the following: a) a polynucleotide sequence encoding a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 11, wherein at least one of the loop residues have been replaced with a variant sequence; and b) a polynucleotide sequence encoding a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 13, wherein at least one of the loop residues have been replaced with a variant sequence.
  • the expression vectors further comprise from the 5′ terminus to the 3′ terminus: a first nucleic acid sequence encoding a signal peptide functional as a secretory sequence in a bacterial cell; a second nucleic acid sequence encoding a secreted polypeptide or functional portion thereof; a third nucleic acid sequence encoding a cleavable linker; and the DNA sequence which encodes the protease inhibitor.
  • the present invention also provides host cells transformed with the expression vectors of the present invention. In some preferred embodiments, the host cell is a Bacillus species cell.
  • the present invention also provides compositions comprising at least one scaffold protein and at least one peptide, wherein the scaffold comprises Bowman-Birk inhibitor.
  • the peptide comprises a peptide that binds to Vet.
  • FIG. 1 provides the DNA and amino acid sequences of the aprE-BCE103-BBI-Histag expression cassette (EcoRI-HindIII) cloned into the pJM103 integration vector (SEQ ID NOS:1 and 2).
  • FIG. 2 provides a schematic map of the pJM103BBIhis expression vector.
  • FIG. 3 provides the DNA and amino acid sequences of 12BBIck81 from the BCE103 fusion site (at the BamHI) to the end of the gene (SEQ ID NOS:3 and 4).
  • the CK37281 peptide sequences (ACYNLYGWTC (SEQ ID NO:9) are inserted into both the trypsin and chymotrypsin inhibitory loops.
  • FIG. 4 provides a graph showing titers of active versus inactive 2BBIck81 (by trypsin inhibition) and the ratio of the activities of BCE103 cellulase to 2BBck81 with various thiol reducing agents added during the growth of the culture.
  • BME 2-mercaptoethanol
  • Cyt cysteine
  • Glut reduced glutathione
  • DIT dithiothreitol
  • FIG. 5 provides a graph showing activation of BCE-lnk2-2BBIck81 with 2-mercaptoethanol (bME) after partial purification by ion exchange chromatography.
  • FIG. 6 provides a graph showing results from a competition analysis of 2BBlck81 versus anti-VegF antibody binding to VegF.
  • FIG. 7 provides the sequence of the synthetic DNA fragment carrying the H. insolens PDI (hiPDI) that was inserted into the B. subtilis BBI expression vector, as well as the amino acid sequence (SEQ ID NOS:5 and 6)
  • FIG. 8 provides the DNA and amino acid sequences of the aprE-cutinase expression cassette that was ligated into the EcoRI-BamHI sites of p2JM103-lnk2-2BBIck81 (SEQ ID NOS:7 and 8).
  • the present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species.
  • the present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • expression cassette and “expression vector” refer to nucleic acid constructs generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell.
  • the recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA virus, or nucleic acid fragment.
  • the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter.
  • expression cassette may be used interchangeably herein with “DNA construct” and its grammatical equivalents.
  • vector and “cloning vector” refer to nucleic acid constructs designed to transfer nucleic acid sequences into cells.
  • expression vector refers to a vector that has the ability to incorporate and express heterologous DNA fragments in a foreign cell.
  • Many prokaryotic and eukaryotic expression vectors are commercially available. Selection of appropriate expression vectors is within the knowledge of those of skill in the art.
  • plasmid refers to a circular double-stranded (ds) DNA construct used as a cloning vector, and which forms an extrachromosomal self-replicating genetic element in some eukaryotes or prokaryotes, or integrates into the host chromosome.
  • nucleic acid molecule and “nucleic acid sequence” include sequences of any form of nucleic acid, including, but not limited to RNA, DNA and cDNA molecules. It will be understood that, as a result of the degeneracy of the genetic code, a multitude of nucleotide sequences encoding a given protein may be produced.
  • a “fusion DNA sequence” comprises from 5′ to 3′ a first, second, third and fourth DNA sequences.
  • first nucleic acid sequence or “first DNA sequence” encodes a signal peptide functional as a secretory sequence in bacterial species.
  • Particularly preferred signal sequences are those derived from polypeptides secreted by the expression host used to express and secrete the fusion polypeptide.
  • first amino acid sequences correspond to secretory sequences which are functional in a bacterial species. Such amino acid sequences are encoded by first DNA sequences as defined.
  • second DNA sequences encode “secreted polypeptides” (i.e., “secreted polypeptides of interest”) expressed by the chosen bacterial species.
  • preferred secreted polypeptides are those which are naturally secreted by the bacterial expression host.
  • heterologous protein i.e., proteins that are not normally secreted by the particular bacterial host.
  • a cellulase expressed by a Bacillus species other than B. subtilis is used that the secreted polypeptide of interest and B. subtilis is used as the expression host.
  • “functional portion of a secreted polypeptide” and its grammatical equivalents refers to a truncated secreted polypeptide that retains its ability to fold into a normal, albeit truncated, configuration. In some embodiments, it is contemplated that sufficient residues of a domain of the secreted polypeptide must be present to allow it to fold in its normal configuration independently of the desired polypeptide to which it is attached. However, in most cases, the portion of the secreted polypeptide are both correctly folded and result in increased secretion as compared to its absence.
  • the truncation of the secreted polypeptide means that the functional portion retains a biological function.
  • the catalytic domain of a secreted polypeptide is used, although other functional domains may be used, for example, the substrate binding domains. Additionally preferred embodiments utilize the catalytic domain and all or part of the linker region.
  • third DNA sequences comprise DNA sequences encoding a cleavable linker polypeptide. It should be understood that the third DNA sequence need only encode that amino acid sequence which is necessary to be recognized by a particular enzyme or chemical agent to bring about cleavage of the fusion polypeptide. Thus, only that portion of the linker which is necessary for recognition and cleavage by the appropriate enzyme is required.
  • fourth DNA sequences encode “desired polypeptides.” Such desired polypeptides include protease inhibitors either in their mature or pro forms, and variants thereof.
  • fusion DNA sequence Such fusion DNA sequences are assembled in proper reading frame from the 5′ terminus to 3′ terminus in the order of first, second, third and fourth DNA sequences.
  • the DNA sequence encodes a “fusion polypeptide,” “fusion protein,” and “fusion analog” encoding from its amino-terminus a signal peptide functional as a secretory sequence in a bacterial species, a secreted polypeptide or portion thereof normally secreted by a bacterial species, a cleavable linker polypeptide and a desired polypeptide.
  • the terms “desired protein” and “desired polypeptide” refer to a polypeptide or protein in its mature or pro form that is not fused to a secretion enhancing construct.
  • a “desired protein” and “desired polypeptide” refer to the protein to be expressed and secreted by the host cell in a non-fused form.
  • fusion polypeptides encode from the amino-terminus a signal peptide functional as a secretory sequence functional in a host cell, a secreted polypeptide or portion thereof normally secreted from a host cell, a cleavable linker polypeptide and a desired polypeptide.
  • the fusion protein is processed by host cell enzymes (e.g., a protease), to yield the desired protein free from the other protein sequences in the fusion protein.
  • host cell enzymes e.g., a protease
  • a “promoter sequence” refers to a DNA sequence which is recognized by the bacterial host for expression purposes. In preferred embodiments, it is operably linked to a DNA sequence encoding the fusion polypeptide. Such linkage comprises positioning of the promoter with respect to the translation initiation codon of the DNA sequence encoding the fusion DNA sequence. In particularly preferred embodiments, the promoter sequence contains transcription and translation control sequences which mediate the expression of the fusion DNA sequence.
  • terminal sequence refers to a DNA sequence which is recognized by the expression host to terminate transcription. It is operably linked to the 3′ end of the fusion DNA encoding the fusion polypeptide to be expressed.
  • selectable marker-encoding nucleotide sequence refers to a nucleotide sequence which is capable of expression in bacterial cells and where expression of the selectable marker confers to cells containing the expressed gene the ability to grow in the presence of a corresponding selective condition.
  • a nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence.
  • DNA encoding a secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide;
  • a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or
  • a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
  • Operably linked DNA sequences are usually contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • recombinant includes reference to a cell or vector, that has been modified by the introduction of a heterologous nucleic acid sequence or that the cell is derived from a cell so modified.
  • recombinant cells express genes that are not found in identical form within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all as a result of deliberate human intervention.
  • the term “expression” refers to the process by which a polypeptide is produced based on the nucleic acid sequence of a gene. The process includes both transcription and translation.
  • the term “protease inhibitor expression” refers to transcription and translation of the specific protease inhibitors and variants thereof gene to be expressed, the products of which include precursor RNA, mRNA, polypeptide, post-translation processed polypeptide, and derivatives thereof.
  • protease inhibitor expression refers to the transcription, translation and assembly of protease inhibitors and variants thereof exemplified by the expression cassette shown in FIG. 1 .
  • assays for protease inhibitor expression include examination of bacterial colonies when exposed to the appropriate conditions, Western blots for protease inhibitor protein, as well as Northern blots analysis and reverse transcriptase polymerase chain reaction (RT-PCR) assays for protease inhibitor mRNA.
  • RT-PCR reverse transcriptase polymerase chain reaction
  • isolated and purified refer to a nucleic acid or polypeptide that is removed from at least one component with which it is naturally associated
  • the term “substantially free” encompasses preparations of the desired polypeptide having less than about 20% (by dry weight) other proteins (i.e., contaminating protein), less than about 10% other proteins, less than about 5% other proteins, or less than about 1% other proteins.
  • the term “substantially pure” when applied to the proteins or fragments thereof of the present invention means that the proteins are essentially free of other substances to an extent practical and appropriate for their intended use.
  • the proteins are sufficiently pure and are sufficiently free from other biological constituents of the host cells so as to be useful in, for example, protein sequencing, and/or producing pharmaceutical preparations.
  • target protein refers to protein (e.g., enzyme, hormone, etc.), whose action would be blocked by the binding of the variant inhibitors provided for herein.
  • variant sequence and “variant sequences” refer to the short polypeptide sequence(s) that replace the binding loops of the wild-type protease inhibitor or other scaffold.
  • the variant sequence does not need to be of the same length as the binding loop sequence it is replacing in the scaffold.
  • the term “scaffold” refers to a wild-type protein sequence into which a variant sequence is introduced.
  • the scaffold has portions (e.g., loops), that are replaced.
  • the BBI sequences provided herein find use as scaffolds for variant sequences.
  • Two protein protease inhibitors have been isolated from soybeans, the Kunitz-type trypsin inhibitor (soybean trypsin inhibitor, STI) and the Bowman-Birk protease inhibitor (BBI) (See e.g., Birk, Int. J. Pept. Protein Res., 25: 113-131 [1985]; and Kennedy, Am. J. Clin. Neutr., 68: 1406 S-1412S [1998]).
  • These inhibitors serve as a scaffold for the variant sequences.
  • other desired proteins used herein include the addition of six histidine residues at the C-terminus (See, FIGS. 1 and 2 ).
  • STI inhibits the proteolytic activity of trypsin by the formation of a stable stoichiometric complex (See e.g., Liu, Chemistry and Nutritional Value of Soybean Components, In: Soybeans, Chemistry, Technology and Utilization , pp. 32-35, Aspen Publishers, Inc., Gaithersburg, Md., [1999]).
  • STI consists of 181 amino acid residues with two disulfide bridges and is roughly spherically shaped (See e.g., Song et al., J. Mol. Biol., 275: 347-63 [1998]).
  • the trypsin inhibitory loop lies within the first disulfide bridge.
  • the Kunitz-type soybean trypsin inhibitor has played a key role in the early study of proteinases, having been used as the main substrate in the biochemical and kinetic work that led to the definition of the standard mechanism of action of proteinase inhibitors.
  • Bowman-Birk inhibitor proteins are a kinetically and structurally well-characterized family of small proteins (60-90 residues) isolated from leguminous seeds, as well as other plants, including various grasses. They typically have a symmetrical structure of two tricyclic domains each containing an independent binding loop, although some have one domain and some have more than two domains.
  • the major 8 kDa Bowman-Birk inhibitor isolated from soybeans (BBI) has two separate reactive site loops, loop I inhibits proteases having trypsin-like specificity and loop II inhibits proteases with chymotrypsin-like specificity (See e.g., Chen et al., J. Biol.
  • binding regions each contain a “canonical loop” structure, which is a motif found in a variety of serine proteinase inhibitors (Bode and Huber, Eur. J. Biochem., 204: 433-451 [1992]). Sn and BBI are found only in the soybean seed, and not in any other part of the plant (See e.g., Birk, Int. J. Pept. Protein Res., 25: 113-131 [1985]).
  • SEQ ID NO:13 shows the amino acid sequence of the BBI backbone used herein comprising approximately 71 amino acid residues (See Example 1).
  • BBI is produced as a pro-protein with an N-terminal pro-peptide that is 19 amino acids in length.
  • BBI is produced with all or at least a portion of the propeptide.
  • BBI is truncated, with as many as 10 amino acid residues being removed from either the N- or C-terminal. For example, upon seed desiccation, some BBI molecules have the C-terminal 9 or 10 amino acid residues removed.
  • proteolysis is generally highly tolerated prior to the initial disulfide and just after the terminal disulfide bond, the consequences of which are usually not detrimental to the binding to target protein.
  • any one of the isoforms or truncated forms find use in various embodiments of the present invention.
  • the STI and BBI protease inhibitors have binding loops that inhibit proteases.
  • the present invention provides protease inhibitor variants with alterations in one or more reactive sites (e.g., Loop I and/or Loop II of BBI).
  • the loops are replaced with sequences that interact with a target protein.
  • the loops are replaced with sequences derived from VEGF binding proteins, inhibitors of the complement pathway such as C2, C3, C4 or C5 inhibitors, Compstatin, cytokines, other proteins of interest, etc.
  • inhibitors of the complement pathway such as C2, C3, C4 or C5 inhibitors, Compstatin, cytokines, other proteins of interest, etc.
  • variant sequences are selected by various methods known in the art, including but not limited to phage display and other suitable screening methods. For example, a random peptide gene library is fused with phage PIII gene so the peptide library will be displayed on the surface of the phage. Subsequently, the phage display library is exposed to the target protein and washed with buffer to remove non-specific binding (this process is sometimes referred to as panning). Finally, the binding phage and PCR the DNA sequence for the peptide encoded are isolated.
  • phage display and other suitable screening methods for example, a random peptide gene library is fused with phage PIII gene so the peptide library will be displayed on the surface of the phage. Subsequently, the phage display library is exposed to the target protein and washed with buffer to remove non-specific binding (this process is sometimes referred to as panning). Finally, the binding phage and PCR the DNA sequence for the peptide encoded are isolated.
  • one of the loops is replaced with a variant sequence (i.e., peptides; often 3 to 14 amino acids in length, with 5 to 10 amino acids being preferred).
  • a variant sequence i.e., peptides; often 3 to 14 amino acids in length, with 5 to 10 amino acids being preferred.
  • Longer sequences find use in the present invention, as long as they provide the binding and/or inhibition desired.
  • peptides suitable for use as replacements of the binding loop(s) preferably adopt a functional conformation when contained within a constrained loop (i.e., a loop formed by the presence of a disulfide bond between two cysteine residues).
  • the peptides are between 7 and 9 amino acids in length.
  • each protease inhibitor or variant thereof is expressed as a fusion protein by the host bacterial cell.
  • cleavage of the fusion polypeptide to release the desired protein will often be useful, it is not necessary.
  • Protease inhibitors and variants thereof expressed and secreted as fusion proteins surprisingly retain their function.
  • fusion DNA sequences are assembled in proper reading frame from the 5′ terminus to 3′ terminus in the order of first, second, third and fourth DNA sequences.
  • the DNA sequence encodes a “fusion polypeptide” encoding from its amino-terminus a signal peptide functional as a secretory sequence in a bacterial species, a secreted polypeptide or portion thereof normally secreted from a bacterial species, a cleavable linker peptide and a desired polypeptide (e.g., a protease inhibitor and variants thereof).
  • the present invention provides bacterial host cells which have been transduced, transformed or transfected with an expression vector comprising a protease inhibitor-encoding nucleic acid sequence.
  • the culture conditions such as temperature, pH and the like, are those previously used for the parental host cell prior to transduction, transformation or transfection are apparent to those skilled in the art.
  • a nucleotide sequence encoding a fusion protein is operably linked to a promoter sequence functional in the host cell.
  • This promoter-gene unit is then typically cloned into intermediate vectors before transformation into the host cells for replication and/or expression.
  • These intermediate vectors are typically prokaryotic vectors (e.g., plasmids, or shuttle vectors).
  • prokaryotic vectors e.g., plasmids, or shuttle vectors.
  • a bacterial culture is transformed with an expression vector having a promoter or biologically active promoter fragment or one or more (e.g., a series) of enhancers which functions in the host cell, operably linked to a nucleic acid sequence encoding a protease inhibitor, such that the a protease is expressed in the cell.
  • the DNA sequences encode a protease inhibitor or variant thereof.
  • the promoter is a regulatable one.
  • Natural or synthetic polynucleotide fragments encoding a protease inhibitor may be incorporated into heterologous nucleic acid constructs or vectors, capable of introduction into, and replication in, a bacterial cell.
  • the vectors and methods disclosed herein are suitable for use in various host cells for the expression of protease inhibitors and variants thereof. Any vector may be used as long as it is replicable and viable in the cells into which it is introduced. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available.
  • vectors Appropriate cloning and expression vectors are also described in various references known to those in the art (See e.g., Sambrook et al., supra and Ausubel et al., supra, expressly incorporated by reference herein).
  • the appropriate DNA sequence is inserted into a plasmid or vector (collectively referred to herein as “vectors”) by any suitable method.
  • vectors a plasmid or vector
  • the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by standard procedures known to those in the art.
  • Appropriate vectors are typically equipped with a selectable marker-encoding nucleic acid sequence, insertion sites, and suitable control elements, such as termination sequences.
  • the vectors comprise regulatory sequences, including, for example, control elements (i.e., promoter and terminator elements or 5′ and/or 3′ untranslated regions), effective for expression of the coding sequence in host cells (and/or in a vector or host cell environment in which a modified soluble protein coding sequence is not normally expressed), operably linked to the coding sequence.
  • control elements i.e., promoter and terminator elements or 5′ and/or 3′ untranslated regions
  • Exemplary promoters include both constitutive promoters and inducible promoters. Such promoters are well known to those of skill in the art. Those skilled in the art are also aware that a natural promoter can be modified by replacement, substitution, addition or elimination of one or more nucleotides without changing its function. The practice of the present invention encompasses and is not constrained by such alterations to the promoter.
  • selectable marker will depend on the host cell. Appropriate markers for different bacterial hosts are well known in the art. Typical selectable marker genes encode proteins that (a) confer resistance to antibiotics or other toxins (e.g., ampicillin, methotrexate, tetracycline, neomycin mycophenolic acid, puromycin, zeomycin, or hygromycin; or (b) complement an auxotrophic mutation or a naturally occurring nutritional deficiency in the host strain.
  • antibiotics or other toxins e.g., ampicillin, methotrexate, tetracycline, neomycin mycophenolic acid, puromycin, zeomycin, or hygromycin.
  • a selected PI coding sequence is inserted into a suitable vector according to well-known recombinant techniques and used to transform a cell line capable of PI expression. Due to the inherent degeneracy of the genetic code, other nucleic acid sequences which encode substantially the same or a functionally equivalent amino acid sequence may be used to clone and express a specific protease inhibitor, as further detailed above. Therefore it is appreciated that such substitutions in the coding region fall within the sequence variants covered by the present invention. Any and all of these sequence variants can be utilized in the same way as described herein for a parent PI-encoding nucleic acid sequence. Those skilled in the art recognize that differing PIs will be encoded by differing nucleic acid sequences.
  • a protease inhibitor nucleic acid sequence, homologue, variant or fragment thereof is obtained, it is modified by any number of ways. Where the sequence involves non-coding flanking regions, the flanking regions may be subjected to resection, mutagenesis, etc. Thus, transitions, transversions, deletions, and insertions may be performed on the naturally occurring sequence.
  • heterologous nucleic acid constructs include the coding sequence for at least one protease inhibitor, or variant(s), fragment(s) or splice variant(s) thereof: (i) in isolation; (ii) in combination with additional coding sequences; such as fusion protein or signal peptide coding sequences, where the PI coding sequence is the dominant coding sequence; (iii) in combination with non-coding sequences, such as control elements, such as promoter and terminator elements or 5′ and/or 3′ untranslated regions, effective for expression of the coding sequence in a suitable host; and/or (iv) in a vector or host environment in which the PI coding sequence is a heterologous gene.
  • heterologous nucleic acid containing the appropriate nucleic acid coding sequence, together with appropriate promoter and control sequences, is employed to introduced into bacterial host cells to permit the cells to express at least one protease inhibitor or variant thereof.
  • a heterologous nucleic acid construct is employed to transfer a PI-encoding nucleic acid sequence into a cell in vitro.
  • the host cells stably integrate the nucleic acid sequences of the present invention.
  • the first and second expression cassettes are present on a single vector, while in other embodiments these cassettes are present on separate vectors.
  • the expression cassette in addition to a promoter sequence, also contains a transcription termination region downstream of the structural gene to provide for efficient termination.
  • the termination region is obtained from the same gene as the promoter sequence, while in other embodiments it is obtained from another gene. The selection of suitable transcription termination signals is well-known to those of skill in the art.
  • any suitable expression vector will find use in the present invention.
  • various conventional vectors used for expression in eukaryotic or prokaryotic cells will be suitable and find use with the present invention.
  • Standard bacterial expression vectors include bacteriophages ⁇ and M13, as well as plasmids such as pBR322 based plasmids, pSKF, pET23D, and fusion expression systems such as MBP, GST, and LacZ.
  • epitope tags are added to recombinant proteins, in order to provide convenient methods of isolation (e.g., c-myc).
  • Additional elements typically included in expression vectors are replicons, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of heterologous sequences.
  • the particular antibiotic resistance gene chosen is not critical, any of the many resistance genes known in the art are suitable.
  • the methods of the present invention provide host cells that contain a stably integrated sequence of interest (i.e., PI-encoding nucleic acid).
  • a stably integrated sequence of interest i.e., PI-encoding nucleic acid.
  • the methods of the present invention provide for maintenance of a self-replicating extra-chromosomal transformation vector.
  • the invention further provides cells and cell compositions which have been genetically modified to comprise an exogenously provided PI-encoding nucleic acid sequence.
  • a parental host cell is genetically modified by an expression vector.
  • the vector is a plasmid, while in other embodiments the vector is a viral particle, phage, naked DNA, etc.
  • the form of the vector be limited to any particular type of vector, as various vectors will find use in the present invention.
  • heterologous nucleic acid constructs comprising a PI-encoding nucleic acid sequence are transcribed in vitro, and the resulting RNA introduced into the host cell by any of the suitable methods known in the art.
  • the genetically modified cells are cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants, and/or amplifying expression of a PI-encoding nucleic acid sequence.
  • the culture conditions such as temperature, pH and the like, are those previously used for the host cell selected for expression, and are apparent to those skilled in the art.
  • the progeny of cells into which such heterologous nucleic acid constructs have been introduced are generally considered to comprise the PI-encoding nucleic acid sequence found in the heterologous nucleic acid construct.
  • Appropriate host cells include any suitable bacterial species.
  • the bacterial hosts serve both as the expression hosts and the source of the first and second nucleic acids.
  • surprising levels of expression have been obtained.
  • the system utilized herein has achieved levels of expression and secretion of greater than 0.5 g/l of protease inhibitor.
  • the transfected host cells are cultured under conditions favoring expression of gene encoding the desired protein. Large batches of transformed cells can be cultured as described above. Finally, product is recovered from the culture using techniques known in the art.
  • Accessory proteins such as thiol-disulfide oxidoreductases or chaperones find use in some embodiments, as they may be beneficial to help fold the secretory protein into its active conformation.
  • Thiol-disulfide oxidoreductases and protein disulfide isomerases catalyze the formation of the correct disulfide bonds in the protein.
  • Overexpression of the bdbDC operon in B. subtilis has been shown to be beneficial for the production of a protein with disulfide bonds (See e.g., Meima et al., J. Biol. Chem., 277: 6994-7001, [2002]).
  • Chaperones help the secretory protein to fold by binding to exposed hydrophobic regions in the unfolded states and preventing unfavourable interactions and prolyl-peptidyl cis-trans isomerases assist in formation of the proper conformation of the peptide chain adjacent to proline residues.
  • the host cells are transformed with an expression vector encoding at least one thiol-disulfide oxidoreductase or chaperone. It is not intended that the present invention be limited to any particular thiol-disulfide oxidoreductase or chaperone, as any suitable thiol-disulfide oxidoreductase or chaperone known to those skilled in the art will find use in the present invention.
  • the fraction of properly folded secretory protein is increased by the addition of chemicals to the growth medium that reduce/oxidize disulfide bonds, and/or alter the general redox potential, and/or chemicals that alter solvent properties thus affecting protein conformation and aggregation.
  • a reagent that reduces disulfide bonds such as 2-mercaptoethanol, is preferred to increase the fraction of correctly folded protein.
  • disulfide reducing or oxidizing agents e.g., DTT, TCEP, reduced and oxidized glutathione, cysteine, cystine, cysteamine, thioglycolate, S 2 O 3 2 ⁇ , S 2 O 4 2 ⁇ , S 2 O 5 2 ⁇ , SO 3 2 ⁇ , S 2 O 7 2 ⁇ , Cu+, etc.
  • DTT disulfide reducing or oxidizing agent
  • TCEP reduced and oxidized glutathione
  • cysteine, cystine, cysteamine, thioglycolate S 2 O 3 2 ⁇ , S 2 O 4 2 ⁇ , S 2 O 5 2 ⁇ , SO 3 2 ⁇ , S 2 O 7 2 ⁇ , Cu+, etc.
  • ⁇ ME di sulfide reducing/oxidizing agents
  • the present invention relies on fermentation procedures for culturing bacterial species. Fermentation procedures for production of heterologous proteins by bacterial species are well known in the art. Culturing is accomplished in a growth medium comprising an aqueous mineral salts mediums organic growth factors, the carbon and energy source material, molecular oxygen (for aerobic and facultative bacteria), and, of course, a starting inoculum of one or more particular microorganism species to be employed.
  • a growth medium comprising an aqueous mineral salts mediums organic growth factors, the carbon and energy source material, molecular oxygen (for aerobic and facultative bacteria), and, of course, a starting inoculum of one or more particular microorganism species to be employed.
  • the media include, in addition to nitrogen, suitable amounts of phosphorus, magnesium, calcium, potassium sulfur, and sodium in suitable soluble assimilable ionic and combined forms, and also present preferably should be certain trace elements such as copper, manganese, molybdenum zinc, iron, boron, and iodine, and others, again in suitable soluble assimilable form, all as known in the art.
  • the fermentation reaction involves an aerobic process in which the molecular oxygen needed is supplied by a molecular oxygen-containing gas such as air, oxygen-enriched air, or even substantially pure molecular oxygen, provided to maintain the contents of the fermentation vessel with a suitable oxygen partial pressure effective in assisting the microorganism species to grow in a fostering fashion.
  • a molecular oxygen-containing gas such as air, oxygen-enriched air, or even substantially pure molecular oxygen
  • aeration generally is conducted at a rate which is in the range of about 0.5 to 10, preferably about 0.5 to 7, volumes (at the pressure employed and at 25° C.) of oxygen-containing gas per liquid volume in the fermentor per minute. This amount is based on air of normal oxygen content being supplied to the reactor, and in terms of pure oxygen the respective ranges would be about 0.1 to 1.7, or preferably about 0.1 to 1.3, volumes (at the pressure employed and at 25° C.) of oxygen per liquid volume in the fermentor per minute.
  • the pressure employed for the microbial conversion process can range widely. Pressures generally are within the range of about 0 to 50 psig, presently preferably about 0 to 30 psig, more preferably at least slightly over atmospheric pressure, as a balance of equipment and operating cost versus oxygen solubility achieved. Greater than atmospheric pressures are advantageous in that such pressures do tend to increase a dissolved oxygen concentration in the aqueous ferment, which in turn can help increase cellular growth rates. At the same time, this is balanced by the fact that high atmospheric pressures do increase equipment and operating costs.
  • the fermentation temperature can vary somewhat, but for most bacterial species used in the present invention, the temperature generally will be within the range of about 20° C. to 40° C., generally preferably in the range of about 28° C. to 37° C., depending on the strain of microorganism chosen, as known to those skilled in the art.
  • the microorganisms also require a source of assimilable nitrogen.
  • the source of assimilable nitrogen can be any nitrogen-containing compound or compounds capable of releasing nitrogen in a form suitable for metabolic utilization by the microorganism. While a variety of organic nitrogen source compounds, such as protein hydrolysates, can be employed, usually cheap nitrogen-containing compounds such as ammonia, ammonium hydroxide, urea, and various ammonium salts such as ammonium phosphate, ammonium sulfate, ammonium pyrophosphate, ammonium chloride, or various other ammonium compounds can be utilized. Ammonia gas itself is convenient for large scale operations, and can be employed by bubbling through the aqueous ferment (fermentation medium) in suitable amounts. At the same time, such ammonia can also be employed to assist in pH control.
  • the pH range in the aqueous microbial ferment should be in the exemplary range of about 2.0 to 8.0.
  • pH range optima for certain microorganisms are dependent on the media employed to some extent, as well as the particular microorganism, and thus change somewhat with change in media as known to those skilled in the art.
  • the fermentation is preferably conducted in such a manner that the carbon-containing substrate can be controlled as a limiting factor, thereby providing good conversion of the carbon-containing substrate to cells and avoiding contamination of the cells with a substantial amount of unconverted substrate.
  • the latter is not a problem with water-soluble substrates, since any remaining traces are readily removed. It may be a problem, however, in the case of non-water-soluble substrates, and require added product-treatment steps such as suitable washing steps.
  • the time needed to reach this limiting substrate level is not critical and may vary with the particular microorganism and fermentation process being conducted. However, it is well known in the art how to determine the carbon source concentration in the fermentation medium and whether or not the desired level of carbon source has been achieved.
  • the fermentation is conducted as a batch or continuous operation
  • fed batch operation is generally preferred for ease of control, production of uniform quantities of products, and most economical uses of all equipment.
  • each of the streams introduced into the reactor preferably is controlled at a predetermined rate, or in response to a need determinable by monitoring such as concentration of the carbon and energy substrate, pH, dissolved oxygen, oxygen or carbon dioxide in the off-gases from the fermentor, cell density measurable by light transmittancy, or the like.
  • the feed rates of the various materials can be varied so as to obtain as rapid a cell growth rate as possible, consistent with efficient utilization of the carbon and energy source, to obtain as high a yield of microorganism cells relative to substrate charge as possible, but more importantly to obtain the highest production of the desired protein per unit volume.
  • all equipment, reactor, or fermentation means, vessel or container, piping, attendant circulating or cooling devices, and the like are initially sterilized, usually by employing steam such as at about 121° C. for at least about 15 minutes.
  • the sterilized reactor then is inoculated with a culture of the selected microorganism in the presence of all the required nutrients, including oxygen, and the carbon-containing substrate.
  • the type of fermentor employed is not critical, though in some embodiments, the 15L Biolafitte (Saint-Gerrnain-en-Laye, France) is preferred.
  • the secreted protein is recovered.
  • the present invention provides methods of separating a desired protein from its fusion analog. It is specifically contemplated that the methods described herein will find use in the separation of proteinase inhibitor and variants from the fusion analog.
  • the collection and purification of the desired protein from the fermentation broth can also be achieved using procedures known to those of skill in the art.
  • the fermentation broth will generally contain cellular debris, including cells, various suspended solids and other biomass contaminants, as well as the desired protein product, which are preferably removed from the fermentation broth by means known in the art. Suitable processes for such removal include conventional solid-liquid separation techniques (e.g., centrifugation, filtration, dialysis, microfiltration, rotary vacuum filtration, or other known processes), to produce a cell-free filtrate.
  • Precipitating the proteinaceous components of the supernatant or filtrate may be accomplished by means of a salt (e.g., ammonium sulfate) or low pH (typically less than 3), followed by purification by a variety of chromatographic procedures (e.g., ion exchange chromatography, affinity chromatography, hydrophobic interaction chromatography, hydrophobic charge induction chromatography etc.) or similar art recognized procedures. It is not intended that the present invention be limited to any particular separation method, as it is contemplated that any method will find use in the present invention.
  • a salt e.g., ammonium sulfate
  • low pH typically less than 3
  • chromatographic procedures e.g., ion exchange chromatography, affinity chromatography, hydrophobic interaction chromatography, hydrophobic charge induction chromatography etc.
  • the polypeptide when the expressed desired polypeptide is secreted from the bacterial cells, the polypeptide is purified from the growth media. In preferred embodiments, the expression host cells are removed from the media before purification of the polypeptide (e.g. by centrifugation).
  • the host cell When the expressed recombinant desired polypeptide is not secreted from the host cell, the host cell is preferably disrupted and the polypeptide released into an aqueous “extract” which is the first stage of purification.
  • the expression host cells are collected from the media before the cell disruption (e.g. by centrifugation).
  • the cell disruption may be performed by using any suitable means known in the art, such as by lysozyme or beta-glucanase digestion or by forcing the cells through high pressure (See e.g., Scobes, Protein Purification , Second edition, Springer-Verlag)
  • the addition of six histidine residues i.e., a “His Tag” to the C-terminus is used as an aid in the purification of the desired protein and its fusion analog.
  • His tags as a purification aid is well known in the art (See e.g., Hengen, TIBS 20: 285-286 [1995]).
  • the 6 ⁇ his-tagged proteins are easily purified using Immobilized Metal ion Affinity Chromatography (IMAC), as known to those skilled in the art.
  • IMAC Immobilized Metal ion Affinity Chromatography
  • the protease inhibitors produced using the present invention be very highly pure (e.g., having a purity of more than 99%). This is particularly true whenever the desired protein is to be used as a therapeutic, but is also necessary for other applications.
  • the methods described herein provide a way of producing substantially pure desired proteins.
  • the desired proteins described herein are useful in pharmaceutical and personal care compositions. However, it is contemplated that proteins of varying purity levels will be produced using the methods of the present invention and it is not intended that the proteins produced using the present invention be limited to any particular level of purity.
  • the activity of the protein is increased by the addition of chemicals that reduce/oxidize disulfide bonds and/or alter the general redox potential, and/or chemicals that alter solvent properties thus affecting protein conformation and aggregation.
  • addition of a reagent that reduces disulfide bonds, such as 2-mercaptoethanol, is used to increase activity of the protein.
  • disulfide reducing or oxidizing agents e.g., DTI, TCEP, reduced and oxidized glutathione, cysteine, cystine, cysteamine, thioglycolate, S 2 O 3 2 ⁇ , S 2 O 4 2 ⁇ , S 2 O 5 2 ⁇ , SO 3 2 ⁇ , S 2 O 7 2 ⁇ , Cu+, protein disulfide-isomerases, protein thiol-disulfide oxidoreductases, etc.), either used alone or in combination, find use in the present invention.
  • Other adjuvants that alter solvent properties e.g.
  • partially purified protein is diluted in buffer (in some particularly preferred embodiments, a zwitterionic buffer with TWEEN®-80 at basic pH) and activated with bME and a disulfide oxidizing agent (in alternative preferred embodiments, oxidized glutathione or sodium sulfite).
  • buffer in some particularly preferred embodiments, a zwitterionic buffer with TWEEN®-80 at basic pH
  • a disulfide oxidizing agent in alternative preferred embodiments, oxidized glutathione or sodium sulfite.
  • conditions will be screened in order to determine the optimal activation of the protein, if desired.
  • various ⁇ ME concentrations 0.1-10 mM
  • oxidizing agent concentrations (0 to 1/20 to 20 times the ⁇ ME concentration) pH (7.5-9.5)
  • temperatures (15-40° C.
  • dilutions (1-20 fold
  • incubation times (12-72 h)
  • aeration incubations under inert gas to vigorous mixing under oxygen containing gases
  • buffer types Tris, CHES, CAPS, Tricine, TAPS, other zwitterionic buffers, etc.
  • buffer concentrations 0.1-1 M
  • various adjuvants known to alter solvent properties thereby affecting protein conformation and aggregation e.g., ethanolamine, DMSO, TWEEN®-80, arginine, urea, etc.
  • PI proteinase inhibitor
  • BBI Boman-Birk inhibitor
  • STI Soybean Trypsin inhibitor
  • VEGF and VegF vascular endothelial growth factor
  • PCR primers used to amplify the BBI gene for fusion to the BCE103 cellulase expression cassette in the pJ103 vector were:
  • BBIfusion_FW (SEQ ID NO:14) 5′ CAGCACGGATCCAGACGATGAGAGCTCTAAACCC 3′
  • BBIHindIII_RV (SEQ ID NO:15) 5′ CTGCAGAAGCTTAAAAATAAAAAAAACGGATTTCCTTCA GGAAATCCGTCCTCTGTTAACTTTTAGTTCTCTTTATCGTCCTCGC 3′
  • BBIHIS-HindIII_RV (SEQ ID NO:16) 5′ CTGCAGAAGCTTAAAAATAAAAAAAAAACGGATT TCCTCAGGAAATCCGTCCTCTGTTAACTTTTAATGGTGATGGTGATGATG GTTCTC 3′
  • FIG. 1 The sequence of the aprE-BCE103-BBI-HisTag expression cassette (EcoRI-HindIII) that was cloned into the pJM103 integration vector is provided in FIG. 1 .
  • FIG. 2 A schematic plasmid map of the pJM103BBIHis expression vector is provided in FIG. 2 .
  • BCE103 alkaline cellulase gene (See, van Soligen, U.S. Pat. No. 6,063,611, hereby incorporated by reference) fused to the B. subtilis aprE promoter and signal sequence, was cloned from pUCAPR103 (Shaw et al., J. Mol.
  • a gene encoding the soybean Bowman-Birk protease inhibitor (BBI) (Swiss-Prot Accession # P01055; See, Odani and Ikenaka, J. Biochem., 71: 839-848 [1972]) with a C-terminal hexa-histidine tag (His-Tag) was synthesized by Operon Technologies (See, DNA sequence above).
  • BBI Bowman-Birk protease inhibitor
  • the BBI gene was amplified by PCR with primers (all primers were synthesized by MWG Biotech, Oligos Etc., or Operon Technologies) that generated a 5′ BamHI site in the correct reading frame with the BCE103 gene, and at the 3′ end introduced a strong transcriptional terminator (LAT, from the Bacillus licheniformis ⁇ -amylase gene) after the end of the BBI gene with a 3′ HindIII site for cloning into the pJM103 vector.
  • LAT strong transcriptional terminator
  • PCR fragments with or without a C-terminal His-Tag were generated with the primers BBIfusion_FW (SEQ ID NO:14) and BBIHISHindIII_RV (SEQ ID NO:16), or BBIfusion_FW (SEQ ID NO:14) and BBI-HindIII_RV (SEQ ID NO:15), respectively, using the synthetic BBI gene as a template.
  • PCR reactions were typically performed on a thermocycler for 30 cycles with High Fidelity Platinum Taq polymerase (Invitrogen) according to the instructions of the supplier (with an annealing temperature of 55° C.).
  • the PCR fragments were cloned as BamHI-HindIII fragments into pJM103 carrying the aprE-BCE103 expression cassette. The correct gene sequence was verified by DNA sequencing.
  • the N-terminus of the mature coding region of the BBI gene was fused in frame to the C-terminal coding region of the first CBD (cellulose binding domain) linker sequence coded by the BCE103 cellulase gene.
  • the two CBD's of BCE103 (Shaw et al., supra) are replaced by BBI in the final expression vectors, pJM103BBI or pJM103BBIhis (See, FIG. 2 ).
  • the aprE promoter controls the expression of the BCE103-BBI gene fusions (See, Ferrari et al., J. Bact., 170: 289-295 [1988]; and Henner et al., J. Bact., 170: 296-300 [1988]).
  • Competent Bacillus subtilis cells BG3934comK (degU Hy 32, oppA, ⁇ spoIIE3501, ⁇ aprE, ⁇ nprE, ⁇ epr, ⁇ ispA, ⁇ bpr, amyE::xylRPxylAcomK-phleo), were transformed with the expression plasmids, pJM103BBI or pJM103BBIhis.
  • the bacteria were made competent by the induction of the comK gene under control of a xylose inducible promoter (Hahn et al., Mol. Microbiol., 21: 763-775 [1996]).
  • the transformants were selected on Luria Broth agar (LA) plates containing 5 ⁇ g/ml chloramphenicol. To increase the expression by gene amplification, colonies were streaked and grown several times on LA plates with 25 ⁇ g/ml chloramphenicol until the growth rate with the antibiotic was similar to growth rate in the absence of chloramphenicol.
  • the BCE103-BBI fusion protein was produced by growth in shake flasks at 37° C. in TSB medium (Tryptone Soya Broth from OXOID, 30 g/L) or in MBD medium, a MOPS based defined medium.
  • MBD medium was made essentially as described (Neidhardt et al., J.
  • micronutrients were made up as a 100 ⁇ stock containing in one liter, 400 mg FeSO 4 .7H 2 O, 100 mg MnSO 4 .H 2 O, 100 mg ZnSO 4 .7H 2 O, 50 mg CuCl 2 .2H 2 O, 100 mg CoCl 2 .6H 2 O, 100 mg NaMoO 4 .2H 2 O, 100 mg Na 2 B 4 O 7 .10H 2 O, 10 ml of 1M CaCl 2 , and 10 ml of 0.5 M sodium citrate.
  • BCE103-BBI fusion protein could be easily visualized in samples from cell free supernatants (after 24 h of growth in TSB medium or 48 h in MBD medium) as the major protein band on SDS-PAGE gels (10% NuPAGE in MES buffer, run as described by the manufacturer, Invitrogen) running at ⁇ 44 kDa by using standard protein stains (e.g. GelCode Blue Stain Reagent; Pierce).
  • the identity of the BCE103-BBI fusion protein was verified by immunoblots of SDS-PAGE gels using the protocols supplied by the manufacturer (BM Chromogenic Western Blotting Kit; Roche Applied Science using an anti-HisTag antibody or an anti-BCE103 cellulase polyclonal antibody for detection).
  • cellulase degradation was assessed qualitatively on LA cellulase indicator plates (with 1% carboxymethylcellulose stained with 0.2% Congo Red, or with 0.5% azo-CM-cellulose, Megazyme), or quantitatively by a direct assay in Assay Buffer (100 mM Tris pH 8.6, 0.005% Tween-80) on the culture broth using a the synthetic substrate, 4-nitrophenyl ⁇ -D-cellobioside (Sigma), using methods known in the art (See e.g., van Tilbeurgh et al., Meth. Enzymol., 160: 45-59 [1988]).
  • Trypsin inhibitory assays were performed in Assay Buffer to determine the BBI activity. Specifically, a standard curve was generated by making eleven 1:1 serial dilutions (100 ⁇ L BBI+100 ⁇ L Assay Buffer) of a 2 ⁇ g/mL standard BBI solution. The BBI standard was purified from a 1 mg/ml Trypsin-Chymotrypsin Inhibitor (Sigma Cat. #T-9777) solution in 20 mM MES pH 6.0 using a hydrophobic interaction column (POROS HP2, Phenyl column, Applied Biosystems).
  • the column was equilibrated with 20 mM MES pH 6.0, loaded with 5 mg of the inhibitor, washed with the equilibration buffer, and then the BBI was eluted with water.
  • Unknown BBI samples to be tested in the inhibitory assay were diluted as necessary, so that two or more data points would fall within the standard curve (usually 1:10, 1:100, 1:200, 1:1000, 1:2000 sample dilutions were tested and then the dilutions fine tuned if necessary).
  • Each diluted BBI standard or sample, 20 ⁇ L was added to 80 ⁇ L of 50 ng/ml bovine pancreatic trypsin (Worthington) (made by diluting a stock 1 mg/mL trypsin solution into Assay Buffer).
  • the standards and samples were arrayed in 96 well microtiter plates.
  • the reactions were mixed and incubated 15 min at 25° C. After the incubation, 100 ⁇ L of the 0.5 mg/ml trypsin substrate (diluted in Assay Buffer from a 100 mg/ml solution in DMSO), Suc-AAPR-pNA (succinyl-Ala-Ala-Pro-Arg-para-nitroanilide, Bachem), was added, mixed and the OD (A 405 ) was monitored for 15 min, with 1 time point recorded every 12 sec using a Spectra Max 250 (Molecular Devices). The data points were used to determine the Vmax for each reaction.
  • Assay Buffer from a 100 mg/ml solution in DMSO
  • Suc-AAPR-pNA succinyl-Ala-Ala-Pro-Arg-para-nitroanilide, Bachem
  • the standard curve was generated by plotting Vmax versus BBI concentration and was fitted to a four-parameter curve. All data fitting was done using software supplied by the manufacturer (Molecular Devices).
  • the BBI concentration of the unknown samples was calculated from the standard curve.
  • the BBI activity was measured using the same protocol but by determining bovine pancreatic chymotrypsin (Worthington) inhibition (chymotrypsin was used at the same concentration as trypsin and chymotrypsin activity was measured by adding 100 ⁇ L of a 0.4 mg/ml chymotrypsin substrate, succinyl-Ala-Ala-Pro-Phe-para-nitroanilide, Bachem).
  • Titers from shake flask runs typically ranged from 0.4-0.9 mg/ml BCE activity and 40-150 ⁇ g/ml BBI trypsin inhibitory activity.
  • titers likely could be improved further by optimizing the bacterial strain, culture medium and growth conditions (aeration, temperature, time of harvest, etc.).
  • fusion proteins to BBI variants and fusion proteins with various linkers between BCE103 and BBI were produced using the methods outlined above, as described in the following Examples.
  • fusion proteins were also produced when the BBI was fused to the 2 nd CBD linker (BCE-cbdD-BBI; See, Example 4) making it possible to use the 1 st CBD to aid in the purification process.
  • 1BBck81+ (SEQ ID NO:19) 5′-CTAAACCCTGTTGCGATCAATGCGCATGTTAATTTGTATGGGTGGAC TTGTCGCTGCAGCGATATGCGTCTG 1BBck81 ⁇ (SEQ ID NO:20) 5′-AATTCAGACGCATATCGCTGCAGCGACAAGTCCACCCATACAAATTA TAACATGCGCATTGATCGCAACAGGGTTTAGAGCT
  • 2BBck81+ (SEQ ID NO:21) 5′-AATTCCTGTCATAGTGCCTGCAAAAGCTGCGCATGTTATAACCTGTA CGGGTGGACCTGTTTTTGCG 2BBck81 ⁇ (SEQ ID NO:22) 5′-TCGACGCAAAAAACAGGTCCACCCGTACAGGTTATAACATGCGCAGCT TTTGCAGGCACTATGACAGG
  • the DNA sequences of the oligonucleotide pairs used to make cassettes to introduce peptides into the trypsin (SacI and EcoRI restriction sites) or chymotypsin (EcoRI and SalI restriction sites) reactive site loops of the synthetic BBI gene are provided below. These peptide coding sequences were then moved into the p2JM103BBI expression vector as SacI-SalI fragments.
  • Comstatin (1 st loop) (SEQ ID NO:23) CTAAACCCTGTTGCGATCAATGCGCATGTGTTGTTCAGGACTGGGGTCAC CACCGTTGTCGCTGCAGCGATATGCGTCTG and (SEQ ID NO:24) AATTCAGACGCATATCGCTGCAGCGACAACGGTGGTGACCCCAGTCCTGA ACAACACATGCGCATTGATCGCAACAGGGTTTAGAGCT Comstatin (2 nd loop) (SEQ ID NO:25) CAAAAGCTGTATCTGCGTTGTTCAGGACTGGGGTCACCACCGTTGTTTTT GCG and (SEQ ID NO:26) TCGACGCAAAAACAACGGTGGTGACCCCAGTCCTGAACAACGCAGATACA GCTTTTGCATG C2c (1 st loop) (SEQ ID NO:27) CTAAACCCTGTTGCGATCAATGCAGCTGTGGTCGTAAAATCCCGATCCAG TGTCGCTGCAGCGATATGCGTCTG and (SEQ ID NO:28) AATTCA
  • the DNA sequences of oligonucleotide primer pairs used to introduce peptide sequences into the trypsin or chymotrypsin reactive site loops using a QuikChange® II XL site-directed mutagenesis kit (Stratagene) are provided below.
  • the reactions were performed as outlined by the manufacturer and described in this Example. Twenty cycles were performed with extensions of 6 minutes at 68° C., denaturations of 50 s at 95° C., and annealings at 55° C. for 50 s. After the cycles, a final extension was performed at 68° C. for 20 minutes.
  • the DNA sequences of the oligonucleotide pair used to make the cassette to introduce the MM021 peptide into the chymotrypsin reactive site loops of the p2JM103-lnk2-BBI expression vector are provided below.
  • the cassette was ligated into the SphI and SalI restriction sites in the vector.
  • cysteine constrained peptides are popular reagents (e.g. the commercially available PhD-C7C Phage Display Peptide Library Kit; NEB) for selecting peptides that bind to substrates of interest.
  • BBI has two cysteine constrained reactive site loops that are structurally similar to the peptide loops displayed in various methods used to select peptide binders. So, once a cysteine constrained binding peptide has been selected, BBI is suitable for use as a scaffold to present the peptide in a binding reaction.
  • the VegF binding peptide CK37281 (See e.g., co-pending U.S. Provisional Patent Application Ser. No. 60/520,403, filed Nov. 13, 2003, incorporated herein by reference) was grafted into BBI by replacing the trypsin, chymotrypsin, or both reactive site loops, with the CK37281 peptide sequence (ACYNLYGWTC) (SEQ ID NO:9) by using DNA oligonucleotide cassettes.
  • an EcoRI site was introduced in the coding region of the BBI gene (custom synthesized by Operon Technologies; See, Example 1) between the trypsin and chymotrypsin reactive site loops by QuikChange® site-directed mutagenesis, using methods described by the manufacturer (Stratagene) using the primers BowBeco-F and BowBeco-R, shown above (0.5 ⁇ mol of each primer was used in the QuikChange® reaction; after an initial denaturation step of 97° C. for 3 minutes, 18 PCR cycles of 68° C. for 12 minutes, 95° C. for 30 seconds and 55° C. for one minute, followed by a final extension reaction for 15 minutes at 68° C.).
  • the CK37281 peptide was grafted into both loops by inserting the CK37281 peptide in the chymotrypsin loop (using the oligonucleotides (2BBck81+ an 2BBck81 ⁇ ) after the trypsin loop was first replaced by the CK37281 peptide.
  • BBI with the CK37281 peptide in the trypsin loop (1BBIck81) was moved into the pJM103BBI expression vector as a SacI-SphI fragment.
  • BBI with the CK37281 in the chymotrypsin loop (2BBIck81), or both loops (12BBIck81) was moved into pJM103BBI as SacI-SalI fragments.
  • the correct sequences were verified by DNA sequencing (the sequence of 12BBIck81 gene is shown in FIG. 3 ).
  • the resulting vectors, pJM103-1BBIck81, pJM103-2BBIck81, or pJM103-12BBIck81, were used to transform B. subtilis BG3934comK, and the production of the BCE fusion proteins was determined as in Example 1 above.
  • the fusion protein running at ⁇ 44 kDa was detected by SDS-PAGE to be the major protein present in the cell free broth. Although in some cases, there was significant degradation (up to 50%) of the BBI moiety (especially after >48 h of growth in MBD medium), as observed by the presence of a prominent protein band running at ⁇ 34 kDa corresponding to the BCE103 catalytic core. In these cases, the titers of the BCE103 cellulase were similar to that measured with fusions to the wild-type BBI (Example 1), but the activity of the BBI (trypsin inhibition with 2BBIck81, or chymotrypsin inhibition with 1BBIck81) was generally about two fold less.
  • a Bacillus subtilis strain with nine protease genes deleted, BG6006 (degU Hy 32, oppA, ⁇ spoIIE3501, ⁇ aprE, ⁇ nprE, ⁇ epr, ⁇ ispA, ⁇ bpr, ⁇ vpr ⁇ wprA, ⁇ mpr-ybjF, ⁇ nprB, amyE::lRPxylAcomK-ermC), was used as an expression host, and the growth temperature (35° C.) and aeration (200 rpm) were reduced. With these changes, a major fusion protein band ( ⁇ 44 kDa) was observed on SDS-PAGE gels with an insignificant band present at the molecular weight expected for the BCE catalytic core protein ( ⁇ 34
  • cysteine constrained peptides were produced when substituted into the trypsin and/or chymotrypsin reactive site loops of BBI fused to the C-terminus of the BCE103 cellulase. Specific examples included:
  • oligonucleotides used to introduce these peptides into either the trypsin (1 st loop) or chymotrypsin (2 nd loop) reactive site loops are provided above.
  • fusion proteins were produced as determined by SDS-PAGE gels.
  • the amount of intact fusion protein was increased by reducing the proteolytic degradation as described above for the CK37281 substituted peptide.
  • the activity of the BBI (by trysin or chymotrypsin inhibition) is typically some 5-20 times lower than what would be expected from the activity of the BCE103 cellulase measured in the cell free supernatants (the two molecules should be present at a 1:1 molar ratio in the fusion protein).
  • An increase in the activity of BBI (measured by either trypsin or chymotrypsin inhibition) in the BCE103-BBI fusion protein can be routinely obtained by adding bME, typically concentrations of 1-4 mM added to the MBD growth medium about 14 h after inoculation.
  • the trypsin or chymotrypsin inhibitory activity of BBI in the fusion protein is also lower than expected when binding peptides (e.g. VegF binding peptide CK37281) replace the chymotrypsin or trypsin reactive site loop, respectively.
  • binding peptides e.g. VegF binding peptide CK37281
  • the inhibitory activity can be increased by treatment with bME.
  • other thiol reducing agents e.g., cysteine, reduced glutathione, DL-dithiothreitol and Tris[2 boxyethyl]phosphine
  • antioxidants e.g., ascorbic acid or DL- ⁇ -tocopherol acetate
  • other adjuvants to the growth medium e.g., isoleucine, soybean oil, Tween-80
  • growth medium e.g., isoleucine, soybean oil, Tween-80
  • cultures of B. subtilis BG6006 transformed with p2JM103-E3-2BBIck81 (See, Example 4, below) were grown in 40 ml MBD medium in 250 ml shake flasks at 37° C. for 13 h. Then, the thiol reducing agents indicated on the graph in FIG. 4 were added and cell supernatants harvested after 62 h of growth. The reagents 2-mercaptoethanol (BME), cysteine (Cys), reduced glutathione (Glut), and DL-dithiothreitol (DTT) were added to the growth medium to the final concentrations indicated on the graph provided in FIG. 4 .
  • BME 2-mercaptoethanol
  • Cys cysteine
  • Glut reduced glutathione
  • DTT DL-dithiothreitol
  • Concentrations of 5 mM ⁇ ME can result in better BCE103:BBI activity ratios but typically result in an overall decrease in both BCE103 and BBI titers (see FIG. 4 ), at least partially due to the reduction in bacterial growth caused by the added reagent.
  • Titers of BCE103 and 2BBIck81 were determined using the assays described in Example 1.
  • BBI activation was also achieved after partial purification of the fusion proteins (e.g. BCE-lnk2-2BBIck81, see Example 4 below) by Q-Sepharose ion exchange chromatography.
  • the fusion protein was purified from cell free broth obtained from shake flasks or fermentor runs. The broth was filtered, diluted five to ten fold in water and the pH adjusted to pH 7.5-8.0. The diluted sample was loaded onto a column packed with Q-Sepharose resin (GE Healthcare). The column was washed with 50 mM Tris pH 7.5 and then washed again in the same buffer containing 300 mM NaCl. The fusion protein was eluted in the same buffer with 700 mM NaCl.
  • the pooled fusion protein fractions were diluted ten fold in Assay Buffer then treated with 2 mM ⁇ ME and 0.2 mM oxidized glutathione (GSSG) with constant mixing on a stir plate or rocker platform for about 24 h at room temperature.
  • the BBI could generally be activated to about 70-100% of the expected trypsin inhibitory activity based on the measured concentration of the BCE103 cellulase.
  • the activation method outlined above generally yielded the best results, in some cases, in order to maximize the activation of a given sample, screens were performed in 96-well plates to determine the optimal conditions.
  • the typical conditions screened were the dilution in Assay Buffer (e.g., a 2-50 fold dilution series), ⁇ ME concentration (e.g., series between 0.5-5 mM) and oxidized glutathione concentration (e.g. 0 mM then a series of 1/20 to 1/2 the ⁇ ME concentration).
  • Assay Buffer e.g., a 2-50 fold dilution series
  • ⁇ ME concentration e.g., series between 0.5-5 mM
  • oxidized glutathione concentration e.g. 0 mM then a series of 1/20 to 1/2 the ⁇ ME concentration
  • the activation of the fusion protein BCE-lnk2-2BBIck81 is shown in FIG. 5 .
  • the fusion protein from a Q-Sepharose purification was diluted 1:10 in Dulbecco's PBS (Mediatech) with 0.005% TWEEN®-80.
  • Beta-mercaptoethanol was added to a final concentration of 3 mM and incubated overnight at room temperature on a rocker. The sample was further incubated at room temperature for about 60 h with vigorous stirring on a magnetic stir plate.
  • the titers of the BCE103 and 2BBIck81 were determined by cellulase assays and trypsin inhibitory assays, respectively.
  • ⁇ ME concentration in the activation reaction it is desirable to reduce the dilution and ⁇ ME concentration in the activation reaction. This can be accomplished by using higher concentrations of buffer (500 mM Tris pH 8.6), or changing to zwitterionic buffers such as CHES (also CAPS, Tricine, TAPS, and other suitable zwitterionic buffers).
  • buffer 500 mM Tris pH 8.6
  • zwitterionic buffers such as CHES (also CAPS, Tricine, TAPS, and other suitable zwitterionic buffers).
  • CHES also CAPS, Tricine, TAPS, and other suitable zwitterionic buffers
  • This Example describes experiments developed to release free BBI or its variants by cleavage of the BCE103-BBI fusion proteins.
  • sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of pJM103-BBI to generate potential cleavage sites during culture growth between the BCE103 catalytic domain and BBI are provided below.
  • BCEsubBBI subtilisin-type sensitive peptide sequence
  • SEQ ID NO:131 GATCCAGGTGGAGCTGCTTTAGTTGACGATGAGAGCT
  • SEQ ID NO:132 CTCATCGTCAACTAAAGCAGCTCCACCTG BCEcbdLBBI (a portion of the 1 st CBD)
  • SEQ ID NO:133 GATCCAGGTGAACCTGACCCAACTCCTCCATCTGATCCTGGAGAATACCC AGCTTGGGACGATGAGAGCT and
  • SEQ ID NO:134 CTCATCGTCCCAAGCTGGGTATTCTCCAGGATCAGATGGAGGAGTTGGGT CAGGTTCACCTG BCEproBBI (the entire pro peptide of BBI) (SEQ ID NO:135) GATCCGGCGAACCTGCGTCTGTCTAAGCTTGGCCTGCTTATGAAATCAGA CCATCAGCACAGCAATGACGATGAGAGCT and (SEQ ID NO:136) CTCATCGTCATTGCTGTGCTGATGGTCTGATT
  • sequences of the DNA oligonucleotide pair that was annealed and ligated into the BamHI and SacI sites of p2JM103-BBI to fuse BBI to the 2 nd CBD linker of BCE103 cellulase are provided below.
  • BCEcbdDBBI (SEQ ID NO:139) GATCCAGGAGAACCGGACCCAACGCCCCCAAGTGATCCAGGAGAGTATCC AGCATGGGATTCAAATCAAATTTACACAAATGAAATTGTGTATCATAACG GTCAGTTATGGCAAGCGAAATGGTGGACACAAAATCAAGAGCCAGGTGAC CCATACGGTCCGTGGGAACCACTCAAATCTGACCCAGATTCAGACGATGA GAGCT and (SEQ ID NO:140) CTCATCGTCTGAATCTGGGTCAGATTTGAGTGGTTCCCACGGACCGTATG GGTCACCTGGCTCTTGATTTTGTGTCCACCATTTCGCTTGCCATAACTGA CCGTTATGATACACAATTTCATTTGTGTAAATTTGATTTGAATCCCATGC TGGATACTCTCCTGGATCACTTGGGGGCGTTGGGTCCGGTTCTCCTG
  • peptide sequences susceptible to acid cleavage between aspartic acid and proline residues are provided below.
  • VVADPN VVADPN (SEQ ID NO:143)(Kempennan et al., Appl. Env. Microbiol., 69: 1589-1597
  • Oligonucleotide primers used to introduce a BssHII site into pJM103BBI by QuikChange® site-directed mutagenesis are provided below.
  • BCEbss-F (SEQ ID NO:144) 5′-TGGCGTTCAGCAACATGAGCGCGCAGGCTGATGATTA BCEbss-R (SEQ ID NO:145) 5′-TAATCATCAGCCTGCGCGCTCATGTTGCTGAACGCCA
  • BCEterm+ (SEQ ID NO:146) 5′-GACATCACGGACTTCTGCTATGAGCCATGTAAACCAAGCGAGGACGA TAAAGAGAACTAAAAGCTTAACTCGAGGTTAACAGAGGACGGATTTCCTG AAGGAAATCCGTTTTTTTATTTTTAATTAAG BCEterm ⁇ (SEQ ID NO:147) 5′-AGCTCTTAATTAAAAATAAAAAAACGGATTTCCTTCAGGAAATCCGT CCTCTGTTAACCTCGAGTTAAGCTTTTAGTTCTCTTTATCGTCCTCGCTT GGTTTACATGGCTCATAGCAGAAGTCCGTGATG
  • PCR primers used to generate the acid labile linkers provided above i.e., Linker 1, Linker 2, and Linker 3
  • Linker 1, Linker 2, and Linker 3 inserted between the BCE103 catalytic domain and BBI are provided below.
  • BCE103coreBssHII_FW (SEQ ID NO:148) 5′-CAGCAACATGAGCGCGCAGGCTG linkerWGDPHY_RV (SEQ ID NO:149) 5′-ATCGTCTGGATCCGGATAGTGGGGGTCTCCCCAAGATGCTGATTCTC TTATTTTTTCCC linkerDNNDPI_RV (SEQ ID NO:150) 5′-ATCGTCTGGATCCGGTATGGGATCATTGTTGTCAGATGCTGATTCTC TTATTTTTTCCC linkerVVADPN_RV (SEQ ID NO:151) 5′-ATCGTCTGGATCCGGGTTGGGATCTGCAACTACAGATGCTGATTCTC TTATTTTTTCCC
  • PCR primers used to generate the acid labile linkers provided above i.e., Linker 1, Linker 2, and Linker 3 inserted into the 1 st CBD linker.
  • Protein sequence of the acid labile linkers inserted between the BCE103 catalytic domain and BBI are provided below.
  • the acid labile linkers are shown in bold type and the sequences from the first CBD domain are underlined.
  • Linker 1 BCE- WGDPHY - PDP -BBI (SEQ ID NO:156) Linker 2 BCE- DNNDPI - PDP -BBI (SEQ ID NO:157) Linker 3 BCE- VVADPN - PDP -BBI (SEQ ID NO:158) LinkerPlus 1 BCE- IPPSDPTPPSDPGEP - WGDPHY - PDP -BBI (SEQ ID NO:159) LinkerPlus 2 BCE- IPPSDPTPPSDPGEP - DNNDPI - PDP -BBI (SEQ ID NO:160) LinkerPlus 3 BCE- IPPSDPTPPSDPGEP - VVADPN - PDP -BBI (SEQ ID NO:161)
  • sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of pJM103-BBI to generate potential cleavage sites between the BCE103 catalytic domain and BBI during the purification process are provided below.
  • BCEentBBI Enteropeptidase cleaveage site
  • SEQ ID NO:162 GATCCAGGTGGAGACGACGATGACAAAGACGATGAGAGCT and (SEQ ID NO:163) CTCATCGTCTTTGTCATCGTCGTCTCCACCTG
  • BCEgenen1BBI Gene I cleavage site
  • SEQ ID NO:164 GATCCAGGTGCTGCTCATTACGACGATGAGAGCT
  • SEQ ID NO:165 CTCATCGTCGTAATGAGCAGCACCTG
  • BCEfurinBBI Furin/Blisterase cleavage site
  • SEQ ID NO:166 GATCCACGTGCTAAAAGAGACGATGAGAGCT and (SEQ ID NO:167) CTCATCGTCTCTTTTAGCACGTG BCEgenen2BBI (Genenase I cleavage site)
  • SEQ ID NO:168 GATCCAGGCGCTGCACACTACAACGACGATGAGAGCT
  • SEQ ID NO:169 CTCATCGTCGTTGTAGTGTGCAGCGCCTG BCEfleBBI (Mpr cleavage site)
  • SEQ ID NO:170 GATCCATTCCTTGAAGACGATGAGAGCT and (SEQ ID NO:171) CTCATCGTCTTCAAGGAATG
  • BCE-Elnk-BBI (Mpr cleavage site) CCCATACCGGAGCCAGACGATGAGAGCTC (SEQ ID NO:172) and CATCGTCTGGCTCCGGTATGGGATCATTGTTG (SEQ ID NO:173)
  • the protein sequence of the E3 linker between the BCE103 catalytic domain and BBI was DNNDPIPEP DDESFNMPIPEP (SEQ ID NO:174).
  • the E Linker is underlined and the sequence generated by faulty recombination in E. coli is shown in bold type. Cleavage by Mpr (or V8 protease) can occur after any of the three glutamic acids present in the E3 Linker.
  • the structure was BCE-(SEQ ID NO:174)-BBI.
  • BCEgenen3BBI GATCCAGGCGCTGCACACTACAAATCAGACCATCAGCACAGCAATGACGATGAGAGCT (SEQ ID NO:175) and CTCATCGTCATTGCTGTGCTGATGGTCTGATTTGTAGTGTGCAGCGCCTG (SEQ ID NO:176)
  • BCEgenen4BBI GATCCAGGCGCTGCACACTACGTAGAATTTCAAGACGATGAGAGCT (SEQ ID NO:177) and CTCATCGTCTTGAAATTCTACGTAGTGTGCAGCGCCTG (SEQ ID NO:178)
  • the protein sequence of a Genenase I sensitive cleavage site (also acid and Mpr sensitive) inserted between the BCE103 catalytic domain and BBI was DNNDPIPDP GAAHYVEFQ (SEQ ID NO:179).
  • the Genenase I site (Gen4 Linker) is in bold type (cleavage occurs between the tyrosine and valine) (NEB) and Linker 2 is underlined. Cleavage by Mpr can also occur after the glutamic acid that follows the valine in the Gen4 linker.
  • the sequence used herein was BCE-SEQ ID NO:179)-BBI.
  • Cleavage sites in the BCE103-lnk2-2BBIck81 fusion protein are indicated below.
  • the C-terminal seven amino acids of the BCE103 catalytic domain underlined
  • linker 2 sequence bold type
  • 2BBIck81 sequences are shown.
  • the acid/heat labile Asp-Pro bonds are indicated with solid headed arrows and the Mpr sensitive bonds after glutamic acids are indicated with line headed arrows.
  • the BBI moiety needs to be cleaved from the BCE103-BBI fusion protein. In some embodiments, this is accomplished during growth, by proteases intrinsically produced by B. subtilis . In some alternative embodiments, this cleavage occurs after growth, during the purification process (e.g. by acid/heat or proteolytic cleavage).
  • Linkers potentially susceptible to cleavage during growth were designed (See, above, sub, cbdL, pro, shortpro, and cbdD) and cloned into the pJM103BBI or p2JM103BBI expression vectors as BamHI-SacI cassettes. The production of fusion protein versus BCE103 catalytic domain was analyzed on SDS-PAGE gels as described in Example 1.
  • the bonds between aspartic acid and proline residues are cleaved by heat treatment at acidic pH as known in the art (See e.g., Landon, Meth. Enzymol., 47: 145-149 [1977]).
  • the 1 st CBD linker in the BCE103 cellulase has three Asp-Pro dipeptide sequences (See, FIG. 1 ) with the potential to be cleaved by acid/heat treatment. However, cleavage by acid/heat treatment at these sites was found to be inefficient.
  • a BssHII site was introduced by QuikChange® XL (Stratagene) mutagenesis (using the manufacturer's methods; and described in Example 2 above, except 8 minute extension and 1 minute denaturation steps were used) in the aprE signal sequence coding region using the oligonucleotide primers BCEbss-F and BCEbss-R (provided above).
  • HindIII and XhoI sites were inserted in front of the LAT terminator (after the BBI stop codon) and a PacI site was added after the terminator (the original HindIII site after the LAT terminator was removed) by inserting an oligonucleotide cassette (BCEterm+ and BCEterm ⁇ ; provided above) into the SalI and the original HindIII sites.
  • This new vector was called “p2JM103BBI.”
  • the acid labile linker fragments were generated by PCR, using forward primer BCE103coreBssHII_FW with each of the reverse primers, linker WGDPHY_R, linker DNNDPI_RV, or linkerVVADPN_RV (the sequences of which are all provided above) and p2JM103BBI as the template (see Example 1 for the PCR protocol).
  • the PCR fragments of 970 bp were digested with BamHI and PstI, the 154 bp fragments encoding the acid linker fragments were isolated from an agarose gel after electrophoresis, and ligated into the p2JM103 vector digested with BamHI and PstI that had also been purified from a gel.
  • Competent B. subtilis strain BG3934comK or BG6006 were transformed with the plasmids, colonies selected on 5 ⁇ g/ml chloramphenicol LA plates and amplified to 25 ⁇ g/ml chloramphenicol as described in Example 1.
  • the acid labile linkers were inserted into the first CBD linker.
  • PCR fragments were generated using the forward primer BCE103corePstI_FW with the reverse primers LplusWGDPHY_RV, LplusDNNDPI_RV, or LplusVVADPN_RV (See above, for the sequences) with p2JM103BBI as a template.
  • the PCR fragments of about 150 bp were digested with BamHI and PstI, purified and ligated to the p2JM103BBI vector digested with BamHI and PstI.
  • the fusion proteins were purified by ion exchange chromatography essentially as described above (See, Example 2).
  • the fusion protein was cleaved by treatment at 55° C. for 16 h in 10% formic acid.
  • the BCE103 catalytic domain precipitated during the acid treatment and was removed by centrifugation.
  • the free BBI in the supernatant was dried overnight on a SpeedVac.
  • the sample was suspended in 50 mM Tris pH 8 before loading on the SDS-PAGE gel.
  • Linker 2 was inserted between the BCE103 catalytic domain and BBI (BCE-DNNDPI-PDP-BBI). This linker was found to be cleaved in a couple of hours at 75° C. in 20 mM glycine pH 2.
  • the fusion protein was cleaved by treatment with a protease during the purification process.
  • Linkers were designed with cleavage sites for glutamic acid specific proteases (e.g., Mpr or V8 protease), Furin/blisterase, Genenase I, and Enteropeptidase (Enterokinase). These linkers were introduced as oligonucleotide cassettes (See above, for the sequences) between the BCE103 catalytic core and BBI in the expression vector using the BamHI and SacI sites (See, FIG. 1 ).
  • the six acid labile linkers described above were tested for cleavage by BsMpr. These fusion proteins were cleaved by treatment for 16 h with 16 ⁇ g of BsMpr at room temperature. After cleavage, the BCE103 catalytic domain was precipitated by the addition of 10% formic acid and removed by centrifugation. The free BBI in the supernatant was dried overnight on a SpeedVac. The sample was suspended in 50 mM Tris pH 8, before loading on the SDS-PAGE. Similar to the acid cleavage, the BCE-DNNDPI-PDP-BBI (Linker 2) fusion protein was much more efficiently cleaved by BsMpr than any of the other linkers.
  • Linker 2 Linker 2
  • BBI and its variants were found to be effectively released from the BCE-DNNDPI-PDP-BBI fusion protein either by acid/heat treatment or proteolytic digestion with a glutamic acid specific protease such as BsMpr.
  • linkers designed for cleaved by Mpr e.g., E, E3 linker, and fle, provided above
  • E, E3 linker, and fle were tested but none of them had any advantages over Linker 2 (the E3 linker was generated by faulty recombination in E. coli after transformation with the QuikChange® site-directed mutagensis reaction designed to construct the E linker).
  • Linkers designed for cleavage by Furin or Blisterase (NEB) BCEfurinBBI
  • Enteropeptidase Entererokinase, NEB
  • BCEentBBI Enteropeptidase
  • Four linkers were also designed (BCEgenen1BBI, BCEgenen2BBI, BCEgenen3BBI, and BCEgenen4BBI) and tested for cleavage by Genenase I (NEB). Efficient cleavage of the fusion protein was observed only with the Gen4 Linker (BCEgenen4BBI).
  • BsMpr was also found to efficiently cleave the Gen4 linker.
  • BsMpr Bacillus licheniformis
  • the BBI is purified away from the BCE103 catalytic domain by selective acid precipitation (pH 3 or lower) of the BCE103 catalytic domain as described above, ion exchange chromatography (See, Example 5), or by selective binding of BBI on an anhydrotrypsin-agarose (Sigma) column loaded in 50 mM Tris pH 8.0, washed with 50 mM Tris pH 8.0 with 150 mM NaCl, then eluting bound BBI with 50 mM glycine pH 2.2 with 300 mM NaCl).
  • the BCE103-lnk2-2BBIck81 fusion protein was produced in B. subtilis as described in Example 2.
  • the fusion protein was purified, and the BBI trypsin inhibitory activity was increased by treatment with ⁇ ME and oxidized glutathione as described in Example 3.
  • the fusion protein was cleaved by BsMpr protease (See, Example 4) and the free 2BBIck81 was purified from the BCE103 catalytic domain by ion exchange chromatography using a Q-Sepharose column.
  • the pH of the cleaved sample was adjusted to 5.5, the sample was th loaded onto the column (equilibrated with 25 mM MES pH 5.5).
  • the free 2BBIck81 was washed through the column using 25 mM sodium acetate pH 5.0 while the BCE103 catalytic core remained bound to the resin.
  • the 2BBIck81 fraction was concentrated by ultrafiltration and analyzed using an electrochemiluminescence (ECL) based binding assay (BioVeris).
  • ECL electrochemiluminescence
  • the Anti-VegF antibody (Santa Cruz) and VegF (PeproTech) were labeled with the electrochemiluminescent dye and biotin, respectively, as described by the manufacturer (BioVeris).
  • Al materials were in Dulbecco's PBS (Mediatech) supplemented with 0.1% TWEEN®-80.
  • An initial dilution series of Anti-VegF antibody (125, 250 and 500 ng/ml) and VegF (100, 150, 200 and 250 ng/ml) were tested in the binding assay to determine the concentrations of each that would give a robust ECL signal.
  • the CK37281 peptide when grafted onto the chymotrypsin inhibitory loop of BBI (2BBIck81) competed with the Anti-VegF antibody for binding to VegF at micromolar concentrations.
  • 2BBIck81 competed for VegF binding better than the synthesized CK37281 peptide itself (See, FIG. 6 ).
  • the CK37281 peptide inserted into the trypsin inhibitory loop, 1BBIck81 also competed with the Anti-VegF antibody in the BioVeris assay.
  • BBI was found to be useful as a scaffold to present active binding peptides selected by various screening methods.
  • oligonucleotide primers used to amplify the dsbC gene ( E. coli ) from pET-40b(+) are provided below. These primers generate a BssHII site at the 5′ end and a BamHI at the 3′ end for cloning into p2JM103-Gen4-2BBIck81.
  • DsbCBBI-F (SEQ ID NO:181) AACATGAGCGCGCAGGCTGATGACGCGGCAATTCAACAAACGTTAG DsbCBBI-R (SEQ ID NO:182) TCGTCTGGATCCGGTATGGGATCATTGTTGTCACCAGAACCACTAGTTGA TCCTTTACCGCTGGTCATTTTTTGGTG
  • CutinaseBBI+ (SEQ ID NO:183) TGCACTTCTCTGCTTTGGTCTGTTGAACGCAGAGGTCTTGACAACAATGA TCCTATTCCG CutinaseBBI ⁇ (SEQ ID NO:184) GATCCGGAATAGGATCATTGTTGTCAAGACCTCTGCGTTCAACAGACCAA AGCAGAGG
  • compositions such as thiol-disulfide oxidoreductases and/or protein disulfide isomerases find use as fusion proteins to help produce correctly folded BBI moieties. In this embodiment, no additional activation step is needed under most circumstances. In additional embodiments, other proteins produced at high titers in B. subtilis also find use as fusion partners.
  • thermostable protein disulfide isomerase from the fungus Humicola insolens has been used as a fusion partner to produce the light chain of immunoglobulin G (2 disulfides) in Bacillus brevis (See, Kajino et al., Appl. Env. Microbiol., 66: 638-642 [2000]).
  • this hiPDI gene was synthesized (DNA2.0) and cloned into the expression vector, p2JM103-lnk2-2BBIck81 (See, Example 4) as a BssHII-SacI fragment.
  • codons occurring with high frequency in highly expressed B. subtilis genes were selected except in cases where restriction sites were introduced or deleted.
  • the N-terminus of the mature hiPDI gene was fused to the AprE signal sequence and the C-terminus was fused to a linker with an Enteropeptidase cleaveage site (Kajino et al., Appl. Env.
  • the production of the PDI-2BBIck81 fusion protein was typically somewhat less than the BCE-2BBck81 grown under identical conditions.
  • the BBI titers (trypsin inhibition) measured from the PDI-2BBIck81 cell free supernatants were also typically less than the BCE-2BBIck81 fusion.
  • the measured activities of BBI when fused to PDI were higher when grown in 2 mM ⁇ ME and the BBI activity was increased by the addition of ⁇ ME to the cell free supernatants after growth when grown in ⁇ ME free medium (as described in Example 3).
  • the thiol-disulfide oxidoreductase activity of PDI does not seem to significantly improve the titers of active 2BBIck81 in the fusion protein or obviate the need for activation of the BBI molecule.
  • DsbC from Escherichia coli was used as a fusion partner for 2BBIck81.
  • the dsbC gene was amplified by PCR using Herculase Enhanced DNA polymerase as described by the manufacturer (Stratagene) using DsbCBBI-F and DsbCBBI-R as primers (sequences shown above) and pET-40b(+) (Novagen) as a template.
  • the isolated PCR fragment was cloned into the vector p2JM103-Gen4-2BBIck81 (See, Example 4) as a BssHII-BamHI fragment.
  • the correct sequence of the fusion gene was verified by DNA sequencing.
  • the titers of the DsbC-2BBIck81 fusion protein were significantly lower than the BCE-2BBIck81 fusion protein as judged on SDS-PAGE gels and the titers of the active 2BBIck81 measured by trypsin inhibition were much lower as well.
  • the aprE-cutinase gene fusion as an EcoRI-Alw44I fragment was ligated with an Alw44I-BamHI linker oligonucleotide cassette (See, sequence above) into the p2JM103-lnk2-2BBIck81 (See, Example 4) that had been cut with EcoRI and BamHI.
  • This cutinase-linker2-2BBkk81 expression vector See, FIG. 8 for the EcoRI-BamHI aprE-cutinase-linker2 sequence
  • the cutinase-linker2-2BBIck81 fusion protein was not the major band observed on SDS-PAGE gels and the measured lipase titers (as measured using the methods provided in U.S. Pat. No. 5,429,950) and BBI titers were much less (ca. 20 fold) than found with the BCE-2BBIck81 fusion protein. Also, the BBI titers in the cutinase fusion protein were not improved significantly when 3 mM ⁇ ME was added to the growth medium. Thus, the highest titers of active 2BBIck81 was consistently obtained by activation of the BCE-2BBIck81 fusion protein. Nonetheless, it is contemplated that various fusion partners will find use in the present invention.

Abstract

The present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species. The present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.

Description

  • The present application claims priority under 35 U.S.C. §119, to co-pending U.S. Provisional Patent Application Ser. No. 60/518,154, filed Nov. 6, 2003, co-pending U.S. Provisional Patent Application Ser. No. 60/520,403, filed Nov. 13, 2003, co-pending U.S. Provisional Patent Application Ser. No. 60/530,954, filed Dec. 19, 2003, co-pending U.S. Provisional Patent Application Ser. No. 60/531,207, filed Dec. 19, 2003, and co-pending U.S. Provisional Patent Application Ser. No. 60/531,189, filed Dec. 19, 2003.
  • FIELD OF THE INVENTION
  • The present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species. The present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • BACKGROUND OF THE INVENTION
  • Proteases are involved in a wide variety of biological processes. Disruption of the balance between proteases and protease inhibitors is often associated with pathologic tissue destruction. Indeed, various studies have focused on the role of proteases in tissue injury, and it is thought that the balance between proteases and protease inhibitors is a major determinant in maintaining tissue integrity. Serine proteases from inflammatory cells, including neutrophils, are implicated in various inflammatory disorders, such as pulmonary emphysema, arthritis, atopic dermatitis and psoriasis.
  • Proteases also appear to function in the spread of certain cancers. Normal cells exist in contact with a complex protein network, called the extracellular matrix (ECM). The ECM is a barrier to cell movement and cancer cells must devise ways to break their attachments, degrade, and move through the ECM in order to metastasize. Proteases are enzymes that degrade other proteins and have long been thought to aid in freeing the tumor cells from their original location by chewing up the ECM. Recent studies have suggested that they may promote cell shape changes and motility through the activation of a protein in the tumor cell membrane called Protease-Activated Receptor-2 (PAR2). This leads to a cascade of intracellular reactions that activates the motility apparatus of the cell. Thus, it is hypothesized that one of the first steps in tumor metastasis is a reorganization of the cell shape, such that it forms a distinct protrusion at one edge facing the direction of migration. The cell then migrates through a blood vessel wall and travels to distal locations, eventually reattaching and forming a metastatic tumor. For example, human prostatic epithelial cells constitutively secrete prostate-specific antigen (PSA), a kallikrein-like serine protease, which is a normal component of the seminal plasma. The protease acts to degrade the extracellular matrix and facilitate invasion of cancerous cells.
  • Synthetic and natural protease inhibitors have been shown to inhibit tumor promotion in vivo and in vitro. Previous investigations have indicated that certain protease inhibitors belonging to a family of structurally-related proteins classified as serine protease inhibitors or SERPINS, are known to inhibit several proteases including trypsin, cathepsin G, thrombin, and tissue kallikrein, as well as neutrophil elastase. The SERPINS are extremely effective at preventing/suppressing carcinogen-induced transformation in vitro and carcinogenesis in animal model systems. Systemic delivery of purified protease inhibitors apparently reduces joint inflammation and cartilage and bone destruction as well.
  • Topical administration of protease inhibitors finds use in such conditions as atopic dermatitis, a common form of inflammation of the skin, which may be localized to a few patches or involve large portions of the body. The depigmenting activity of protease inhibitors and their capability to prevent ultraviolet-induced pigmentation have been demonstrated both in vitro and in vivo (See e.g., Paine et al., J. Invest. Dermatol., 116: 587-595 [2001]). Protease inhibitors have also been reported to facilitate wound healing. For example, secretory leukocyte protease inhibitor was demonstrated to reverse the tissue destruction and speed the wound healing process when topically applied. In addition, serine protease inhibitors can also help to reduce pain in lupus erythematosus patients (See e.g., U.S. Pat. No. 6,537,968).
  • As noted above, protease inhibitors interfere with the action of proteases. Naturally occurring protease inhibitors can be found in a variety of foods such as cereal grains (oats, barley, and maize), Brussels sprouts, onion, beetroot, wheat, finger millet, and peanuts. One source of interest is the soybean. The average level of protease inhibitors present in soybeans is around 1.4 percent and 0.6 percent for Kunitz and Bowman-Birk respectively, two of the most important protease inhibitors. Notably, these low levels make it impractical to isolate the natural protease inhibitor for clinical applications.
  • Thus, there is a need for methods and compositions suitable for the large-scale production of protease inhibitors and their variants. In particular, there remains a need for compositions and methods that reduce and/or eliminate risks associated with blood-borne infectious agents when these proteases are produced in mammalian tissue culture cells.
  • SUMMARY OF THE INVENTION
  • The present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species. The present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • Provided herein are nucleic acids, cells and methods for the production of protease inhibitors and variants thereof.
  • The present invention provides nucleic acids encoding at least one functional protease inhibitor. In one aspect, a nucleic acid comprising regulatory sequences operatively linked to a first, second, third and fourth nucleic acid sequences are provided. In some embodiments, terminator sequences are provided following the fourth nucleic acid sequence.
  • In alternative embodiments, the first nucleic acid sequence encodes a signal polypeptide functional as a secretory sequence in a first host organism, the second nucleic acid encodes a secreted polypeptide or functional portion thereof normally secreted from the first or a second host organism, the third nucleic acid encodes a cleavable linker and the fourth nucleic acid encodes a protease inhibitor or fragment thereof. In further embodiments, the present invention provides at least one expression cassette comprising nucleic acid sequences encoding at least one protease inhibitor.
  • In additional embodiments, the present invention provides polynucleotides encoding at least one protease inhibitor variant. In some particularly preferred embodiments, the polynucleotide encodes a Bowman-Birk Inhibitor (BBI) variant, wherein at least one loop of the wild-type BBI has been altered.
  • The present invention also provides methods of expressing functional protease inhibitors or variants thereof. In some preferred embodiments, host cells suitable for production of functional protease inhibitors and/or variants thereof are provided. In some embodiments, a host cell is (i) transformed with at least one expression cassette comprising a nucleic acid sequence encoding at least one protease inhibitor or variant thereof, and (ii) cultured under appropriate conditions to express at least one protease inhibitor or variants thereof. In some embodiments of the methods, the method further comprises recovering the protease inhibitor or variant thereof.
  • In alternative embodiments, a host cell is (i) transformed with a first expression cassette comprising at least one nucleic acid sequence encoding a protease inhibitor or variant thereof, (ii) transformed with a second expression cassette comprising a nucleic acid sequence encoding at least one thiol-disulfide oxidoreductase or chaperone, and (iii) cultured under appropriate conditions to express the protease inhibitors or variant thereof. In some embodiments, the protease inhibitors or variants thereof are recovered. In some preferred embodiments, the protease inhibitors or variant thereof are expressed as a fusion protein. In further embodiments, the methods further comprise recovering the protease inhibitor or variant thereof.
  • In still further embodiments, functional protease inhibitors and variants thereof are provided. In some embodiments, the functional protease inhibitor or variant thereof is expressed as a fusion protein consisting of the signal sequence, a cellulase catalytic domain, a cleavable linker region, and then by the mature protease inhibitor or variant thereof.
  • In additional embodiments, the expressed proteins are treated with a protease and/or acid/heat treatment to liberate a protease inhibitor or variant thereof from the fusion protein.
  • In further embodiments, the present invention provides a polypeptide having protease inhibitory activity, selected from the group consisting of
  • a) Bowman-Birk Inhibitor variants;
  • b) Bowman-Birk Inhibitor, and
  • c) A scaffold comprising at least one variant sequence.
  • The present invention provides compositions comprising a fusion protein, wherein the fusion protein comprises a protease inhibitor, and a peptide of interest. In some embodiments, the fusion protein comprises an amino acid selected from the group consisting of SEQ ID NOS:2, 4, 6 and 8. In alternative embodiments, the fusion protein is encoded by a nucleotide sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, and 7. In some preferred embodiments, the protease inhibitor is selected from the group consisting of Bowman-Birk inhibitor (BBI), soybean trypsin inhibitor (STI), and eglin C. In some particularly preferred embodiments, the protease inhibitor is BBI, and wherein the BBI comprises at least one loop selected from the group consisting of a trypsin loop and a chymotrypsin loop. In further preferred embodiments, the protease inhibitor is a scaffold for the peptide of interest. In some preferred embodiments, the loop comprises at least one peptide of interest. In some particularly preferred embodiments, the peptide of interest comprises the amino acid sequence set forth in SEQ ID NO:9. In some more preferred embodiments, the fusion protein comprises the amino acid sequence set forth in SEQ ID NO:4.
  • The present invention also provides methods for producing at least one protease inhibitor in a bacterial cell comprising: a) introducing a DNA construct into a bacterial cell, wherein the DNA construct comprises a heterologous DNA sequence encoding a protease inhibitor derived from a Bowman-Birk Inhibitor (BBI) or variants thereof; b) culturing the bacterial cell under suitable culture conditions to allow expression of the heterologous DNA sequence; and c) producing the protease inhibitor. In some embodiments, the construct further comprises at least one catalytic domain. In some embodiments, catalytic domain is selected from the group consisting of cellulase, cutinase, and disulfide isomerase. In some preferred embodiments, the method further comprises the step of recovering the protease inhibitor. In some particularly preferred embodiments, the method further comprises the step of activating the protease inhibitor. In some most preferred embodiments, the activating is accomplished by exposing the protease inhibitor to at least one reagent selected from the group consisting of compositions that reduce disulfide bonds, compositions that oxidize disulfide bonds, and compositions that alter the redox potential. In some embodiments, the bacterial cell is a member of the genus Bacillus. In additional embodiments, protease inhibitor is selected from the group consisting of: i) a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 11; and ii) a protease inhibitor having at least 90% sequence identity with SEQ ID NO:13. In yet further embodiments, the protease inhibitor comprises a variant sequence. In additional embodiments, the protease inhibitor is Bowman-Birk inhibitor and wherein SEQ ID NO:9 is substituted for at least one loop, wherein the loop is selected from the group consisting of the trypsin loop and the chymotrypsin loop. In some embodiments, the method further comprises the step of introducing a second nucleic acid sequence encoding a thiol-disulfide oxidoreductase or chaperone into the bacterial cell. In some preferred embodiments, the protease inhibitor is expressed as a fusion protein. In some preferred embodiments, the fusion protein further comprises a cellulase catalytic domain, a cleavage site, and the protease inhibitor. In some particularly preferred embodiments, the fusion protein is processed by a protease or acid/heat treatment to liberate the protease inhibitor. In still further preferred embodiments, the fusion protein further comprises at least one linker sequence. In some embodiments, the linker sequence is selected from the group consisting of SEQ ID NOS:141-143.
  • The present invention also provides protease inhibitor compositions comprising the protease inhibitor produced according to any and all of the methods described herein. In some embodiments, the various primers and oligonucleotides described herein find use in the production of the fusion proteins of the present invention. In addition, the present invention provides numerous peptides suitable for use in the present invention.
  • The present invention also provides methods for inhibiting the proteolytic activity of a target protein comprising contacting the target protein with any of the protease inhibitor compositions described herein, and binding the target protein wherein the proteolytic activity of the target protein i inhibited.
  • The present invention further provides isolated polynucleotides encoding a protease inhibitor selected from the group consisting of polypeptide sequences set forth in SEQ ID NOS:10 and 12.
  • The present invention also provides expression vectors comprising a polynucleotide sequence, wherein the polynucleotide sequence is selected from the following: a) a polynucleotide sequence encoding a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 11, wherein at least one of the loop residues have been replaced with a variant sequence; and b) a polynucleotide sequence encoding a protease inhibitor having at least 90% sequence identity with SEQ ID NO: 13, wherein at least one of the loop residues have been replaced with a variant sequence. In some embodiments, the expression vectors further comprise from the 5′ terminus to the 3′ terminus: a first nucleic acid sequence encoding a signal peptide functional as a secretory sequence in a bacterial cell; a second nucleic acid sequence encoding a secreted polypeptide or functional portion thereof; a third nucleic acid sequence encoding a cleavable linker; and the DNA sequence which encodes the protease inhibitor. The present invention also provides host cells transformed with the expression vectors of the present invention. In some preferred embodiments, the host cell is a Bacillus species cell.
  • The present invention also provides compositions comprising at least one scaffold protein and at least one peptide, wherein the scaffold comprises Bowman-Birk inhibitor. In some preferred embodiments, the peptide comprises a peptide that binds to Vet.
  • Other objects, features and advantages of the present invention are apparent from the following detailed description. It should be understood, however, that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the scope and spirit of the invention will be apparent to those skilled in the art from this detailed description.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 provides the DNA and amino acid sequences of the aprE-BCE103-BBI-Histag expression cassette (EcoRI-HindIII) cloned into the pJM103 integration vector (SEQ ID NOS:1 and 2).
  • FIG. 2 provides a schematic map of the pJM103BBIhis expression vector.
  • FIG. 3 provides the DNA and amino acid sequences of 12BBIck81 from the BCE103 fusion site (at the BamHI) to the end of the gene (SEQ ID NOS:3 and 4). The CK37281 peptide sequences (ACYNLYGWTC (SEQ ID NO:9) are inserted into both the trypsin and chymotrypsin inhibitory loops.
  • FIG. 4 provides a graph showing titers of active versus inactive 2BBIck81 (by trypsin inhibition) and the ratio of the activities of BCE103 cellulase to 2BBck81 with various thiol reducing agents added during the growth of the culture. In this Figure, BME=2-mercaptoethanol, Cyt=cysteine, Glut=reduced glutathione, DIT=dithiothreitol).
  • FIG. 5 provides a graph showing activation of BCE-lnk2-2BBIck81 with 2-mercaptoethanol (bME) after partial purification by ion exchange chromatography.
  • FIG. 6 provides a graph showing results from a competition analysis of 2BBlck81 versus anti-VegF antibody binding to VegF.
  • FIG. 7 provides the sequence of the synthetic DNA fragment carrying the H. insolens PDI (hiPDI) that was inserted into the B. subtilis BBI expression vector, as well as the amino acid sequence (SEQ ID NOS:5 and 6)
  • FIG. 8 provides the DNA and amino acid sequences of the aprE-cutinase expression cassette that was ligated into the EcoRI-BamHI sites of p2JM103-lnk2-2BBIck81 (SEQ ID NOS:7 and 8).
  • DESCRIPTION OF THE INVENTION
  • The present invention provides compositions and methods related to expression of protease inhibitors and variants thereof in bacterial species. The present invention further provides fusion nucleic acids, vectors, fusion polypeptides, and processes for obtaining the protease inhibitors.
  • Unless otherwise indicated, the practice of the present invention involves conventional techniques commonly used in molecular biology, microbiology, and recombinant DNA, which are within the skill of the art. Such techniques are known to those of skill in the art and are described in numerous texts and reference works (See e.g., Sambrook et al., “Molecular Cloning: A Laboratory Manual”, Second Edition (Cold Spring Harbor), [1989]); and Ausubel et al., “Current Protocols in Molecular Biology” [1987]). All patents, patent applications, articles and publications mentioned herein, both supra and infra, are hereby expressly incorporated herein by reference.
  • Unless defined otherwise herein, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. For example, Singleton and Sainsbury, Dictionary of Microbiology and Molecular Biology, 2d Ed., John Wiley and Sons, NY (1994); and Hale and Marham, The Harper Collins Dictionary of Biology, Harper Perennial, N.Y. (1991) provide those of skill in the art with a general dictionaries of many of the terms used in the invention. Although any methods and materials similar or equivalent to those described herein find use in the practice of the present invention, the preferred methods and materials are described herein. Accordingly, the terms defined immediately below are more fully described by reference to the Specification as a whole. Also, as used herein, the singular “a”, “an” and “the” includes the plural reference unless the context clearly indicates otherwise. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, nucleic acids are written left to right in 5′ to 3′ orientation; amino acid sequences are written left to right in amino to carboxy orientation, respectively. It is to be understood that this invention is not limited to the particular methodology, protocols, and reagents described, as these may vary, depending upon the context they are used by those of skill in the art.
  • Furthermore, the headings provided herein are not limitations of the various aspects or embodiments of the invention which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification as a whole. Nonetheless, in order to facilitate understanding of the invention, a number of terms are defined below.
  • DEFINITIONS
  • As used herein, the terms “expression cassette” and “expression vector” refer to nucleic acid constructs generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a target cell. The recombinant expression cassette can be incorporated into a plasmid, chromosome, mitochondrial DNA, plastid DNA virus, or nucleic acid fragment. Typically, the recombinant expression cassette portion of an expression vector includes, among other sequences, a nucleic acid sequence to be transcribed and a promoter. The term “expression cassette” may be used interchangeably herein with “DNA construct” and its grammatical equivalents.
  • As used herein, the terms “vector” and “cloning vector” refer to nucleic acid constructs designed to transfer nucleic acid sequences into cells.
  • As used herein, the term “expression vector” refers to a vector that has the ability to incorporate and express heterologous DNA fragments in a foreign cell. Many prokaryotic and eukaryotic expression vectors are commercially available. Selection of appropriate expression vectors is within the knowledge of those of skill in the art.
  • As used herein, the term “plasmid” refers to a circular double-stranded (ds) DNA construct used as a cloning vector, and which forms an extrachromosomal self-replicating genetic element in some eukaryotes or prokaryotes, or integrates into the host chromosome.
  • As used herein, the terms “nucleic acid molecule” and “nucleic acid sequence” include sequences of any form of nucleic acid, including, but not limited to RNA, DNA and cDNA molecules. It will be understood that, as a result of the degeneracy of the genetic code, a multitude of nucleotide sequences encoding a given protein may be produced.
  • As used herein, a “fusion DNA sequence” comprises from 5′ to 3′ a first, second, third and fourth DNA sequences.
  • As used herein, “a first nucleic acid sequence” or “first DNA sequence” encodes a signal peptide functional as a secretory sequence in bacterial species. Particularly preferred signal sequences are those derived from polypeptides secreted by the expression host used to express and secrete the fusion polypeptide. As used herein, first amino acid sequences correspond to secretory sequences which are functional in a bacterial species. Such amino acid sequences are encoded by first DNA sequences as defined.
  • As used herein, “second DNA sequences” encode “secreted polypeptides” (i.e., “secreted polypeptides of interest”) expressed by the chosen bacterial species. As with the first DNA sequences, preferred secreted polypeptides are those which are naturally secreted by the bacterial expression host. However, in some embodiments, the term refers to heterologous protein (i.e., proteins that are not normally secreted by the particular bacterial host). For example, in some preferred embodiments, a cellulase expressed by a Bacillus species other than B. subtilis is used that the secreted polypeptide of interest and B. subtilis is used as the expression host.
  • As used herein, “functional portion of a secreted polypeptide” and its grammatical equivalents refers to a truncated secreted polypeptide that retains its ability to fold into a normal, albeit truncated, configuration. In some embodiments, it is contemplated that sufficient residues of a domain of the secreted polypeptide must be present to allow it to fold in its normal configuration independently of the desired polypeptide to which it is attached. However, in most cases, the portion of the secreted polypeptide are both correctly folded and result in increased secretion as compared to its absence.
  • Similarly, in most cases, the truncation of the secreted polypeptide means that the functional portion retains a biological function. In a preferred embodiment, the catalytic domain of a secreted polypeptide is used, although other functional domains may be used, for example, the substrate binding domains. Additionally preferred embodiments utilize the catalytic domain and all or part of the linker region.
  • As used herein, “third DNA sequences” comprise DNA sequences encoding a cleavable linker polypeptide. It should be understood that the third DNA sequence need only encode that amino acid sequence which is necessary to be recognized by a particular enzyme or chemical agent to bring about cleavage of the fusion polypeptide. Thus, only that portion of the linker which is necessary for recognition and cleavage by the appropriate enzyme is required.
  • As used herein, “fourth DNA sequences” encode “desired polypeptides.” Such desired polypeptides include protease inhibitors either in their mature or pro forms, and variants thereof.
  • The above-defined four DNA sequences encoding the corresponding four amino acid sequences are combined to form a “fusion DNA sequence.” Such fusion DNA sequences are assembled in proper reading frame from the 5′ terminus to 3′ terminus in the order of first, second, third and fourth DNA sequences. As so assembled, the DNA sequence encodes a “fusion polypeptide,” “fusion protein,” and “fusion analog” encoding from its amino-terminus a signal peptide functional as a secretory sequence in a bacterial species, a secreted polypeptide or portion thereof normally secreted by a bacterial species, a cleavable linker polypeptide and a desired polypeptide.
  • As used herein, the terms “desired protein” and “desired polypeptide” refer to a polypeptide or protein in its mature or pro form that is not fused to a secretion enhancing construct. Thus, a “desired protein” and “desired polypeptide” refer to the protein to be expressed and secreted by the host cell in a non-fused form.
  • As used herein, “fusion polypeptides,” “fusion proteins,” and “fusion analogs” encode from the amino-terminus a signal peptide functional as a secretory sequence functional in a host cell, a secreted polypeptide or portion thereof normally secreted from a host cell, a cleavable linker polypeptide and a desired polypeptide. In some embodiments, the fusion protein is processed by host cell enzymes (e.g., a protease), to yield the desired protein free from the other protein sequences in the fusion protein. As used herein, the terms “fusion analog,” “fusion polypeptide,” and “fusion protein” are used interchangeably.
  • As used herein, a “promoter sequence” refers to a DNA sequence which is recognized by the bacterial host for expression purposes. In preferred embodiments, it is operably linked to a DNA sequence encoding the fusion polypeptide. Such linkage comprises positioning of the promoter with respect to the translation initiation codon of the DNA sequence encoding the fusion DNA sequence. In particularly preferred embodiments, the promoter sequence contains transcription and translation control sequences which mediate the expression of the fusion DNA sequence.
  • As used herein, “terminator sequence” refers to a DNA sequence which is recognized by the expression host to terminate transcription. It is operably linked to the 3′ end of the fusion DNA encoding the fusion polypeptide to be expressed.
  • As used herein, the term “selectable marker-encoding nucleotide sequence” refers to a nucleotide sequence which is capable of expression in bacterial cells and where expression of the selectable marker confers to cells containing the expressed gene the ability to grow in the presence of a corresponding selective condition.
  • As used herein, a nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For example, DNA encoding a secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. Operably linked DNA sequences are usually contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restriction sites. If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice.
  • As used herein, “recombinant” includes reference to a cell or vector, that has been modified by the introduction of a heterologous nucleic acid sequence or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found in identical form within the native (non-recombinant) form of the cell or express native genes that are otherwise abnormally expressed, under expressed or not expressed at all as a result of deliberate human intervention.
  • As used herein, the term “expression” refers to the process by which a polypeptide is produced based on the nucleic acid sequence of a gene. The process includes both transcription and translation. Thus, the term “protease inhibitor expression” refers to transcription and translation of the specific protease inhibitors and variants thereof gene to be expressed, the products of which include precursor RNA, mRNA, polypeptide, post-translation processed polypeptide, and derivatives thereof. Similarly, “protease inhibitor expression” refers to the transcription, translation and assembly of protease inhibitors and variants thereof exemplified by the expression cassette shown in FIG. 1. By way of example, assays for protease inhibitor expression include examination of bacterial colonies when exposed to the appropriate conditions, Western blots for protease inhibitor protein, as well as Northern blots analysis and reverse transcriptase polymerase chain reaction (RT-PCR) assays for protease inhibitor mRNA.
  • As used herein, the terms “isolated” and “purified” as used herein refer to a nucleic acid or polypeptide that is removed from at least one component with which it is naturally associated
  • As used herein, the term “substantially free” encompasses preparations of the desired polypeptide having less than about 20% (by dry weight) other proteins (i.e., contaminating protein), less than about 10% other proteins, less than about 5% other proteins, or less than about 1% other proteins.
  • As used herein, the term “substantially pure” when applied to the proteins or fragments thereof of the present invention means that the proteins are essentially free of other substances to an extent practical and appropriate for their intended use. In particular, the proteins are sufficiently pure and are sufficiently free from other biological constituents of the host cells so as to be useful in, for example, protein sequencing, and/or producing pharmaceutical preparations.
  • As used herein, the term “target protein” refers to protein (e.g., enzyme, hormone, etc.), whose action would be blocked by the binding of the variant inhibitors provided for herein.
  • As used herein, the terms “variant sequence” and “variant sequences” refer to the short polypeptide sequence(s) that replace the binding loops of the wild-type protease inhibitor or other scaffold. The variant sequence does not need to be of the same length as the binding loop sequence it is replacing in the scaffold.
  • As used herein, the term “scaffold” refers to a wild-type protein sequence into which a variant sequence is introduced. In some embodiments, the scaffold has portions (e.g., loops), that are replaced. For example, the BBI sequences provided herein find use as scaffolds for variant sequences.
  • Protease Inhibitors
  • Two protein protease inhibitors have been isolated from soybeans, the Kunitz-type trypsin inhibitor (soybean trypsin inhibitor, STI) and the Bowman-Birk protease inhibitor (BBI) (See e.g., Birk, Int. J. Pept. Protein Res., 25: 113-131 [1985]; and Kennedy, Am. J. Clin. Neutr., 68: 1406 S-1412S [1998]). These inhibitors serve as a scaffold for the variant sequences. In addition to alterations in the scaffold comprising the variant sequences, other desired proteins used herein include the addition of six histidine residues at the C-terminus (See, FIGS. 1 and 2).
  • Soybean Trypsin Inhibitor (STI)
  • STI inhibits the proteolytic activity of trypsin by the formation of a stable stoichiometric complex (See e.g., Liu, Chemistry and Nutritional Value of Soybean Components, In: Soybeans, Chemistry, Technology and Utilization, pp. 32-35, Aspen Publishers, Inc., Gaithersburg, Md., [1999]). STI consists of 181 amino acid residues with two disulfide bridges and is roughly spherically shaped (See e.g., Song et al., J. Mol. Biol., 275: 347-63 [1998]). The trypsin inhibitory loop lies within the first disulfide bridge. The Kunitz-type soybean trypsin inhibitor (STI) has played a key role in the early study of proteinases, having been used as the main substrate in the biochemical and kinetic work that led to the definition of the standard mechanism of action of proteinase inhibitors.
  • Bowman-Birk Inhibitor (BBI)
  • Bowman-Birk inhibitor proteins are a kinetically and structurally well-characterized family of small proteins (60-90 residues) isolated from leguminous seeds, as well as other plants, including various grasses. They typically have a symmetrical structure of two tricyclic domains each containing an independent binding loop, although some have one domain and some have more than two domains. The major 8 kDa Bowman-Birk inhibitor isolated from soybeans (BBI) has two separate reactive site loops, loop I inhibits proteases having trypsin-like specificity and loop II inhibits proteases with chymotrypsin-like specificity (See e.g., Chen et al., J. Biol. Chem., 267: 1990-1994 [1992]; Werner and Wemmer, Biochem., 31: 999-1010 [1992]; Lin et al., Eur. J. Biochem., 212: 549-555 [1993]; Voss et al., Eur. J. Biochem., 242: 122-131 [1996]; and Billings et al., Pro. Natl. Acad. Sci., 89: 3120-3124 [1992]). These binding regions each contain a “canonical loop” structure, which is a motif found in a variety of serine proteinase inhibitors (Bode and Huber, Eur. J. Biochem., 204: 433-451 [1992]). Sn and BBI are found only in the soybean seed, and not in any other part of the plant (See e.g., Birk, Int. J. Pept. Protein Res., 25: 113-131 [1985]).
  • Although numerous isoforms of BBI have been characterized, SEQ ID NO:13 shows the amino acid sequence of the BBI backbone used herein comprising approximately 71 amino acid residues (See Example 1).
  • In soybeans, BBI is produced as a pro-protein with an N-terminal pro-peptide that is 19 amino acids in length. Thus, in some embodiments, BBI is produced with all or at least a portion of the propeptide. In some embodiments, BBI is truncated, with as many as 10 amino acid residues being removed from either the N- or C-terminal. For example, upon seed desiccation, some BBI molecules have the C-terminal 9 or 10 amino acid residues removed. Thus, proteolysis is generally highly tolerated prior to the initial disulfide and just after the terminal disulfide bond, the consequences of which are usually not detrimental to the binding to target protein. However, it will be appreciated that any one of the isoforms or truncated forms find use in various embodiments of the present invention.
  • Protease Inhibitor Variants
  • As indicated above, the STI and BBI protease inhibitors have binding loops that inhibit proteases. The present invention provides protease inhibitor variants with alterations in one or more reactive sites (e.g., Loop I and/or Loop II of BBI). In some preferred embodiments, the loops are replaced with sequences that interact with a target protein.
  • For example, in some embodiments, the loops are replaced with sequences derived from VEGF binding proteins, inhibitors of the complement pathway such as C2, C3, C4 or C5 inhibitors, Compstatin, cytokines, other proteins of interest, etc. Indeed, it is not intended that the present invention be limited to any particular sequence substituted into either of these loops, as any suitable sequence finds use in the present invention.
  • In some embodiments, variant sequences are selected by various methods known in the art, including but not limited to phage display and other suitable screening methods. For example, a random peptide gene library is fused with phage PIII gene so the peptide library will be displayed on the surface of the phage. Subsequently, the phage display library is exposed to the target protein and washed with buffer to remove non-specific binding (this process is sometimes referred to as panning). Finally, the binding phage and PCR the DNA sequence for the peptide encoded are isolated.
  • In most embodiments, one of the loops is replaced with a variant sequence (i.e., peptides; often 3 to 14 amino acids in length, with 5 to 10 amino acids being preferred). Longer sequences find use in the present invention, as long as they provide the binding and/or inhibition desired. In addition, peptides suitable for use as replacements of the binding loop(s) preferably adopt a functional conformation when contained within a constrained loop (i.e., a loop formed by the presence of a disulfide bond between two cysteine residues). In some specific embodiments, the peptides are between 7 and 9 amino acids in length. These replacement sequences also provide protease inhibition or binding to the targeted proteins. In some embodiments, it is advantages to alter a single amino acid.
  • Fusion Proteins
  • In preferred embodiments, each protease inhibitor or variant thereof is expressed as a fusion protein by the host bacterial cell. Although cleavage of the fusion polypeptide to release the desired protein will often be useful, it is not necessary. Protease inhibitors and variants thereof expressed and secreted as fusion proteins surprisingly retain their function.
  • The above-defined DNA sequences encoding the corresponding amino acid sequences are combined to form a “fusion DNA sequence.” Such fusion DNA sequences are assembled in proper reading frame from the 5′ terminus to 3′ terminus in the order of first, second, third and fourth DNA sequences. As so assembled, the DNA sequence encodes a “fusion polypeptide” encoding from its amino-terminus a signal peptide functional as a secretory sequence in a bacterial species, a secreted polypeptide or portion thereof normally secreted from a bacterial species, a cleavable linker peptide and a desired polypeptide (e.g., a protease inhibitor and variants thereof). Various methods are known to those in the art for the production of fusion proteins (See e.g., U.S. Pat. Nos. 5,411,873, 5,429,950, and 5,679,543, all of which are incorporated by reference herein). Thus, it is intended that any suitable method will find use in the present invention.
  • Expression of Recombinant Protease Inhibitors
  • To the extent that the present invention depends on the production of fusion proteins, it relies on routine techniques in the field of recombinant genetics. Basic texts disclosing the general methods of use in this invention include Sambrook et al., Molecular Cloning, A Laboratory Manual ((2nd ed.) [1989]); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Ausubel et al., (eds.), Current Protocols in Molecular Biology (1994).
  • The present invention provides bacterial host cells which have been transduced, transformed or transfected with an expression vector comprising a protease inhibitor-encoding nucleic acid sequence. The culture conditions, such as temperature, pH and the like, are those previously used for the parental host cell prior to transduction, transformation or transfection are apparent to those skilled in the art.
  • Basically, a nucleotide sequence encoding a fusion protein is operably linked to a promoter sequence functional in the host cell. This promoter-gene unit is then typically cloned into intermediate vectors before transformation into the host cells for replication and/or expression. These intermediate vectors are typically prokaryotic vectors (e.g., plasmids, or shuttle vectors). However, it is not intended that the present invention be limited to the use of intermediate vectors, as this step is omitted in some preferred embodiments.
  • In one approach, a bacterial culture is transformed with an expression vector having a promoter or biologically active promoter fragment or one or more (e.g., a series) of enhancers which functions in the host cell, operably linked to a nucleic acid sequence encoding a protease inhibitor, such that the a protease is expressed in the cell. In some preferred embodiments, the DNA sequences encode a protease inhibitor or variant thereof. In another preferred embodiment, the promoter is a regulatable one.
  • Nucleic Acid Constructs/Expression Vectors.
  • Natural or synthetic polynucleotide fragments encoding a protease inhibitor (i.e., “PI-encoding nucleic acid sequences”) may be incorporated into heterologous nucleic acid constructs or vectors, capable of introduction into, and replication in, a bacterial cell. The vectors and methods disclosed herein are suitable for use in various host cells for the expression of protease inhibitors and variants thereof. Any vector may be used as long as it is replicable and viable in the cells into which it is introduced. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. Appropriate cloning and expression vectors are also described in various references known to those in the art (See e.g., Sambrook et al., supra and Ausubel et al., supra, expressly incorporated by reference herein). The appropriate DNA sequence is inserted into a plasmid or vector (collectively referred to herein as “vectors”) by any suitable method. In general, the DNA sequence is inserted into an appropriate restriction endonuclease site(s) by standard procedures known to those in the art.
  • Appropriate vectors are typically equipped with a selectable marker-encoding nucleic acid sequence, insertion sites, and suitable control elements, such as termination sequences. In some embodiments, the vectors comprise regulatory sequences, including, for example, control elements (i.e., promoter and terminator elements or 5′ and/or 3′ untranslated regions), effective for expression of the coding sequence in host cells (and/or in a vector or host cell environment in which a modified soluble protein coding sequence is not normally expressed), operably linked to the coding sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, many of which are commercially available and known to those in the art.
  • Exemplary promoters include both constitutive promoters and inducible promoters. Such promoters are well known to those of skill in the art. Those skilled in the art are also aware that a natural promoter can be modified by replacement, substitution, addition or elimination of one or more nucleotides without changing its function. The practice of the present invention encompasses and is not constrained by such alterations to the promoter.
  • The choice of promoter used in the genetic construct is within the knowledge of one skilled in the art.
  • The choice of the proper selectable marker will depend on the host cell. Appropriate markers for different bacterial hosts are well known in the art. Typical selectable marker genes encode proteins that (a) confer resistance to antibiotics or other toxins (e.g., ampicillin, methotrexate, tetracycline, neomycin mycophenolic acid, puromycin, zeomycin, or hygromycin; or (b) complement an auxotrophic mutation or a naturally occurring nutritional deficiency in the host strain.
  • In some embodiments, a selected PI coding sequence is inserted into a suitable vector according to well-known recombinant techniques and used to transform a cell line capable of PI expression. Due to the inherent degeneracy of the genetic code, other nucleic acid sequences which encode substantially the same or a functionally equivalent amino acid sequence may be used to clone and express a specific protease inhibitor, as further detailed above. Therefore it is appreciated that such substitutions in the coding region fall within the sequence variants covered by the present invention. Any and all of these sequence variants can be utilized in the same way as described herein for a parent PI-encoding nucleic acid sequence. Those skilled in the art recognize that differing PIs will be encoded by differing nucleic acid sequences.
  • In some embodiments, once the desired form of a protease inhibitor nucleic acid sequence, homologue, variant or fragment thereof, is obtained, it is modified by any number of ways. Where the sequence involves non-coding flanking regions, the flanking regions may be subjected to resection, mutagenesis, etc. Thus, transitions, transversions, deletions, and insertions may be performed on the naturally occurring sequence.
  • In some preferred embodiments, heterologous nucleic acid constructs include the coding sequence for at least one protease inhibitor, or variant(s), fragment(s) or splice variant(s) thereof: (i) in isolation; (ii) in combination with additional coding sequences; such as fusion protein or signal peptide coding sequences, where the PI coding sequence is the dominant coding sequence; (iii) in combination with non-coding sequences, such as control elements, such as promoter and terminator elements or 5′ and/or 3′ untranslated regions, effective for expression of the coding sequence in a suitable host; and/or (iv) in a vector or host environment in which the PI coding sequence is a heterologous gene.
  • In some embodiments, heterologous nucleic acid containing the appropriate nucleic acid coding sequence, together with appropriate promoter and control sequences, is employed to introduced into bacterial host cells to permit the cells to express at least one protease inhibitor or variant thereof.
  • In some embodiments of the present invention, a heterologous nucleic acid construct is employed to transfer a PI-encoding nucleic acid sequence into a cell in vitro. In some preferred embodiments, the host cells stably integrate the nucleic acid sequences of the present invention. Thus, any suitable method for effectively generating stable transformants finds use in the present invention.
  • In additional embodiments of the present invention, the first and second expression cassettes are present on a single vector, while in other embodiments these cassettes are present on separate vectors.
  • In some preferred embodiments, in addition to a promoter sequence, the expression cassette also contains a transcription termination region downstream of the structural gene to provide for efficient termination. In some embodiments, the termination region is obtained from the same gene as the promoter sequence, while in other embodiments it is obtained from another gene. The selection of suitable transcription termination signals is well-known to those of skill in the art.
  • In addition, it is contemplated that any suitable expression vector will find use in the present invention. Indeed, it is contemplated that various conventional vectors used for expression in eukaryotic or prokaryotic cells will be suitable and find use with the present invention. Standard bacterial expression vectors include bacteriophages λ and M13, as well as plasmids such as pBR322 based plasmids, pSKF, pET23D, and fusion expression systems such as MBP, GST, and LacZ. In further embodiments, epitope tags are added to recombinant proteins, in order to provide convenient methods of isolation (e.g., c-myc).
  • Additional elements typically included in expression vectors are replicons, a gene encoding antibiotic resistance to permit selection of bacteria that harbor recombinant plasmids, and unique restriction sites in nonessential regions of the plasmid to allow insertion of heterologous sequences. The particular antibiotic resistance gene chosen is not critical, any of the many resistance genes known in the art are suitable.
  • Introduction of a Protease Inhibitor-Encoding Nucleic Acid Sequence Into Host Cells.
  • In some preferred embodiments, the methods of the present invention provide host cells that contain a stably integrated sequence of interest (i.e., PI-encoding nucleic acid). However, in alternative embodiments, the methods of the present invention provide for maintenance of a self-replicating extra-chromosomal transformation vector.
  • The invention further provides cells and cell compositions which have been genetically modified to comprise an exogenously provided PI-encoding nucleic acid sequence. In some embodiments, a parental host cell is genetically modified by an expression vector. In some embodiments, the vector is a plasmid, while in other embodiments the vector is a viral particle, phage, naked DNA, etc. Thus, it is not intended that the form of the vector be limited to any particular type of vector, as various vectors will find use in the present invention.
  • Various methods may be employed for delivering an expression vector into cells in vitro. Methods of introducing nucleic acids into cells for expression of heterologous nucleic acid sequences are also known to the ordinarily skilled artisan, including, but not limited to electroporation; protoplast fusion with intact cells; transduction; high velocity bombardment with DNA-coated microprojectiles; infection with modified viral (e.g., phage) nucleic acids; chemically-mediated transformation, competence, etc. In addition, in some embodiments, heterologous nucleic acid constructs comprising a PI-encoding nucleic acid sequence are transcribed in vitro, and the resulting RNA introduced into the host cell by any of the suitable methods known in the art.
  • Following introduction of a heterologous nucleic acid construct comprising the coding sequence for a protease inhibitor, the genetically modified cells are cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants, and/or amplifying expression of a PI-encoding nucleic acid sequence. The culture conditions, such as temperature, pH and the like, are those previously used for the host cell selected for expression, and are apparent to those skilled in the art.
  • The progeny of cells into which such heterologous nucleic acid constructs have been introduced are generally considered to comprise the PI-encoding nucleic acid sequence found in the heterologous nucleic acid construct.
  • Bacterial Hosts and Expression
  • Appropriate host cells include any suitable bacterial species. In some embodiments, the bacterial hosts serve both as the expression hosts and the source of the first and second nucleic acids. Using the present inventive methods and host cells, surprising levels of expression have been obtained. The system utilized herein has achieved levels of expression and secretion of greater than 0.5 g/l of protease inhibitor.
  • After the expression vector is introduced into the host cells, the transfected host cells are cultured under conditions favoring expression of gene encoding the desired protein. Large batches of transformed cells can be cultured as described above. Finally, product is recovered from the culture using techniques known in the art.
  • Accessory proteins such as thiol-disulfide oxidoreductases or chaperones find use in some embodiments, as they may be beneficial to help fold the secretory protein into its active conformation. Thiol-disulfide oxidoreductases and protein disulfide isomerases catalyze the formation of the correct disulfide bonds in the protein. Overexpression of the bdbDC operon in B. subtilis has been shown to be beneficial for the production of a protein with disulfide bonds (See e.g., Meima et al., J. Biol. Chem., 277: 6994-7001, [2002]). Chaperones help the secretory protein to fold by binding to exposed hydrophobic regions in the unfolded states and preventing unfavourable interactions and prolyl-peptidyl cis-trans isomerases assist in formation of the proper conformation of the peptide chain adjacent to proline residues.
  • In some embodiments of the present invention, the host cells are transformed with an expression vector encoding at least one thiol-disulfide oxidoreductase or chaperone. It is not intended that the present invention be limited to any particular thiol-disulfide oxidoreductase or chaperone, as any suitable thiol-disulfide oxidoreductase or chaperone known to those skilled in the art will find use in the present invention.
  • In some embodiments of the present invention, the fraction of properly folded secretory protein is increased by the addition of chemicals to the growth medium that reduce/oxidize disulfide bonds, and/or alter the general redox potential, and/or chemicals that alter solvent properties thus affecting protein conformation and aggregation. In particularly preferred embodiments, a reagent that reduces disulfide bonds, such as 2-mercaptoethanol, is preferred to increase the fraction of correctly folded protein. However, in other embodiments and depending on the medium used, other disulfide reducing or oxidizing agents (e.g., DTT, TCEP, reduced and oxidized glutathione, cysteine, cystine, cysteamine, thioglycolate, S2O3 2−, S2O4 2−, S2O5 2−, SO3 2−, S2O7 2−, Cu+, etc.), either used alone or in combination, find use in the present invention. It is contemplated that other adjuvants that alter solvent properties, (e.g., urea, DMSO, TWEEN®-80, etc.), either added to the growth medium alone or preferably in combination with di sulfide reducing/oxidizing agents, such as βME, will also increase the fraction of correctly folded secretory protein and find use in various embodiments of the present invention. In some preferred embodiments, the βME is used at concentrations ranging from 0.5 to 4 mM, while in other embodiments, the concentrations range from 0.1 mM to 10 mM. Indeed, those of skill in the art know how to select the best growth medium and growth conditions to optimize the effects of the added thiol reducing/oxidizing agents and/or other adjuvants, as well as the concentration of thio reducing/oxidizing agents and/or other adjuvants to use. It is not intended that the present invention be limited to any particular disulfide reducing/oxidizing agent or adjuvant, as any suitable reagents known to those skilled in the art find use in the present invention.
  • Fermentation Parameters
  • The present invention relies on fermentation procedures for culturing bacterial species. Fermentation procedures for production of heterologous proteins by bacterial species are well known in the art. Culturing is accomplished in a growth medium comprising an aqueous mineral salts mediums organic growth factors, the carbon and energy source material, molecular oxygen (for aerobic and facultative bacteria), and, of course, a starting inoculum of one or more particular microorganism species to be employed.
  • In addition to the carbon and energy source, oxygen, assimilable nitrogen, and an inoculum of the microorganism, it is necessary to supply suitable amounts in proper proportions of mineral nutrients to assure proper microorganism growth, maximize the assimilation of the carbon and energy source by the cells in the microbial conversion process, and achieve maximum cellular yields with maximum cell density in the fermentation medium.
  • Various culture media find use in the present invention, as known to those of skill in the art. However, standard bacterial culture media find use in the present invention. In some preferred media formulations, the media include, in addition to nitrogen, suitable amounts of phosphorus, magnesium, calcium, potassium sulfur, and sodium in suitable soluble assimilable ionic and combined forms, and also present preferably should be certain trace elements such as copper, manganese, molybdenum zinc, iron, boron, and iodine, and others, again in suitable soluble assimilable form, all as known in the art.
  • In some embodiments, the fermentation reaction involves an aerobic process in which the molecular oxygen needed is supplied by a molecular oxygen-containing gas such as air, oxygen-enriched air, or even substantially pure molecular oxygen, provided to maintain the contents of the fermentation vessel with a suitable oxygen partial pressure effective in assisting the microorganism species to grow in a thriving fashion. In effect, by using an oxygenated hydrocarbon substrate, the oxygen requirement for growth of the microorganism is reduced. Nevertheless, molecular oxygen must be supplied for growth of aerobic and to a lesser extent, facultative organisms.
  • Although the aeration rate can vary over a considerable range, aeration generally is conducted at a rate which is in the range of about 0.5 to 10, preferably about 0.5 to 7, volumes (at the pressure employed and at 25° C.) of oxygen-containing gas per liquid volume in the fermentor per minute. This amount is based on air of normal oxygen content being supplied to the reactor, and in terms of pure oxygen the respective ranges would be about 0.1 to 1.7, or preferably about 0.1 to 1.3, volumes (at the pressure employed and at 25° C.) of oxygen per liquid volume in the fermentor per minute.
  • The pressure employed for the microbial conversion process can range widely. Pressures generally are within the range of about 0 to 50 psig, presently preferably about 0 to 30 psig, more preferably at least slightly over atmospheric pressure, as a balance of equipment and operating cost versus oxygen solubility achieved. Greater than atmospheric pressures are advantageous in that such pressures do tend to increase a dissolved oxygen concentration in the aqueous ferment, which in turn can help increase cellular growth rates. At the same time, this is balanced by the fact that high atmospheric pressures do increase equipment and operating costs.
  • The fermentation temperature can vary somewhat, but for most bacterial species used in the present invention, the temperature generally will be within the range of about 20° C. to 40° C., generally preferably in the range of about 28° C. to 37° C., depending on the strain of microorganism chosen, as known to those skilled in the art.
  • The microorganisms also require a source of assimilable nitrogen. The source of assimilable nitrogen can be any nitrogen-containing compound or compounds capable of releasing nitrogen in a form suitable for metabolic utilization by the microorganism. While a variety of organic nitrogen source compounds, such as protein hydrolysates, can be employed, usually cheap nitrogen-containing compounds such as ammonia, ammonium hydroxide, urea, and various ammonium salts such as ammonium phosphate, ammonium sulfate, ammonium pyrophosphate, ammonium chloride, or various other ammonium compounds can be utilized. Ammonia gas itself is convenient for large scale operations, and can be employed by bubbling through the aqueous ferment (fermentation medium) in suitable amounts. At the same time, such ammonia can also be employed to assist in pH control.
  • The pH range in the aqueous microbial ferment (fermentation admixture) should be in the exemplary range of about 2.0 to 8.0. However, pH range optima for certain microorganisms are dependent on the media employed to some extent, as well as the particular microorganism, and thus change somewhat with change in media as known to those skilled in the art.
  • While the average retention time of the fermentation admixture in the fermentor can vary considerably, depending in part on the fermentation temperature and culture employed, as known in the art.
  • In some embodiments, the fermentation is preferably conducted in such a manner that the carbon-containing substrate can be controlled as a limiting factor, thereby providing good conversion of the carbon-containing substrate to cells and avoiding contamination of the cells with a substantial amount of unconverted substrate. The latter is not a problem with water-soluble substrates, since any remaining traces are readily removed. It may be a problem, however, in the case of non-water-soluble substrates, and require added product-treatment steps such as suitable washing steps. The time needed to reach this limiting substrate level is not critical and may vary with the particular microorganism and fermentation process being conducted. However, it is well known in the art how to determine the carbon source concentration in the fermentation medium and whether or not the desired level of carbon source has been achieved.
  • Although in some embodiments, the fermentation is conducted as a batch or continuous operation, fed batch operation is generally preferred for ease of control, production of uniform quantities of products, and most economical uses of all equipment.
  • If desired, part or all of the carbon and energy source material and/or part of the assimilable nitrogen source such as ammonia can be added to the aqueous mineral medium prior to feeding the aqueous mineral medium into the fermentor. Indeed, each of the streams introduced into the reactor preferably is controlled at a predetermined rate, or in response to a need determinable by monitoring such as concentration of the carbon and energy substrate, pH, dissolved oxygen, oxygen or carbon dioxide in the off-gases from the fermentor, cell density measurable by light transmittancy, or the like. The feed rates of the various materials can be varied so as to obtain as rapid a cell growth rate as possible, consistent with efficient utilization of the carbon and energy source, to obtain as high a yield of microorganism cells relative to substrate charge as possible, but more importantly to obtain the highest production of the desired protein per unit volume.
  • In either a batch, or the preferred fed batch operation, all equipment, reactor, or fermentation means, vessel or container, piping, attendant circulating or cooling devices, and the like, are initially sterilized, usually by employing steam such as at about 121° C. for at least about 15 minutes. The sterilized reactor then is inoculated with a culture of the selected microorganism in the presence of all the required nutrients, including oxygen, and the carbon-containing substrate. The type of fermentor employed is not critical, though in some embodiments, the 15L Biolafitte (Saint-Gerrnain-en-Laye, France) is preferred.
  • Protein Separations
  • In preferred embodiments, once the desired protein is expressed, the secreted protein is recovered. The present invention provides methods of separating a desired protein from its fusion analog. It is specifically contemplated that the methods described herein will find use in the separation of proteinase inhibitor and variants from the fusion analog.
  • The collection and purification of the desired protein from the fermentation broth can also be achieved using procedures known to those of skill in the art. The fermentation broth will generally contain cellular debris, including cells, various suspended solids and other biomass contaminants, as well as the desired protein product, which are preferably removed from the fermentation broth by means known in the art. Suitable processes for such removal include conventional solid-liquid separation techniques (e.g., centrifugation, filtration, dialysis, microfiltration, rotary vacuum filtration, or other known processes), to produce a cell-free filtrate. In some embodiments, it is preferable to further concentrate the fermentation broth or the cell-free filtrate prior to the purification and/or crystallization process using techniques such as ultrafiltration, evaporation and/or precipitation.
  • Precipitating the proteinaceous components of the supernatant or filtrate may be accomplished by means of a salt (e.g., ammonium sulfate) or low pH (typically less than 3), followed by purification by a variety of chromatographic procedures (e.g., ion exchange chromatography, affinity chromatography, hydrophobic interaction chromatography, hydrophobic charge induction chromatography etc.) or similar art recognized procedures. It is not intended that the present invention be limited to any particular separation method, as it is contemplated that any method will find use in the present invention.
  • In certain preferred embodiments, when the expressed desired polypeptide is secreted from the bacterial cells, the polypeptide is purified from the growth media. In preferred embodiments, the expression host cells are removed from the media before purification of the polypeptide (e.g. by centrifugation).
  • When the expressed recombinant desired polypeptide is not secreted from the host cell, the host cell is preferably disrupted and the polypeptide released into an aqueous “extract” which is the first stage of purification. Preferably, the expression host cells are collected from the media before the cell disruption (e.g. by centrifugation). The cell disruption may be performed by using any suitable means known in the art, such as by lysozyme or beta-glucanase digestion or by forcing the cells through high pressure (See e.g., Scobes, Protein Purification, Second edition, Springer-Verlag)
  • In some embodiments, the addition of six histidine residues (i.e., a “His Tag”) to the C-terminus is used as an aid in the purification of the desired protein and its fusion analog. Use of the His tags as a purification aid is well known in the art (See e.g., Hengen, TIBS 20: 285-286 [1995]). The 6×his-tagged proteins are easily purified using Immobilized Metal ion Affinity Chromatography (IMAC), as known to those skilled in the art.
  • Purity
  • For some applications, it is of great importance that the protease inhibitors produced using the present invention be very highly pure (e.g., having a purity of more than 99%). This is particularly true whenever the desired protein is to be used as a therapeutic, but is also necessary for other applications. The methods described herein provide a way of producing substantially pure desired proteins. The desired proteins described herein are useful in pharmaceutical and personal care compositions. However, it is contemplated that proteins of varying purity levels will be produced using the methods of the present invention and it is not intended that the proteins produced using the present invention be limited to any particular level of purity.
  • Activation of BBI During Purification
  • In some embodiments of the present invention, after growth during the purification process, the activity of the protein is increased by the addition of chemicals that reduce/oxidize disulfide bonds and/or alter the general redox potential, and/or chemicals that alter solvent properties thus affecting protein conformation and aggregation. In some particularly preferred embodiments, addition of a reagent that reduces disulfide bonds, such as 2-mercaptoethanol, is used to increase activity of the protein. However, as those skilled in the art appreciate, depending purity and buffer composition, other disulfide reducing or oxidizing agents (e.g., DTI, TCEP, reduced and oxidized glutathione, cysteine, cystine, cysteamine, thioglycolate, S2O3 2−, S2O4 2−, S2O5 2−, SO3 2−, S2O7 2−, Cu+, protein disulfide-isomerases, protein thiol-disulfide oxidoreductases, etc.), either used alone or in combination, find use in the present invention. Other adjuvants that alter solvent properties, (e.g. ethanolamine, DMSO, TWEEN®-80, arginine, urea, etc.), either added alone or preferably in combination with disulfide reducing/oxidizing agents, such as βME, during the purification process also find use in the present invention by increasing the activity of the protein. In certain preferred embodiments, partially purified protein is diluted in buffer (in some particularly preferred embodiments, a zwitterionic buffer with TWEEN®-80 at basic pH) and activated with bME and a disulfide oxidizing agent (in alternative preferred embodiments, oxidized glutathione or sodium sulfite).
  • In addition, it is contemplated that conditions will be screened in order to determine the optimal activation of the protein, if desired. For example, various βME concentrations (0.1-10 mM), oxidizing agent concentrations (0 to 1/20 to 20 times the βME concentration) pH (7.5-9.5), temperatures (15-40° C.), dilutions (1-20 fold), incubation times (12-72 h), aeration (incubations under inert gas to vigorous mixing under oxygen containing gases), buffer types (Tris, CHES, CAPS, Tricine, TAPS, other zwitterionic buffers, etc.), buffer concentrations (0.1-1 M), and the addition of various adjuvants known to alter solvent properties thereby affecting protein conformation and aggregation (e.g., ethanolamine, DMSO, TWEEN®-80, arginine, urea, etc.) are tested in order to determine the optimum conditions for the expression system used. It is not intended that the present invention be limited to any particular disulfide reducing/oxidizing agent, dilution, temperature, pH, buffer type or composition, or adjuvant, as any suitable reagents known to those skilled in the art find use in the present invention.
  • EXPERIMENTAL
  • The present invention is described in further detail in the following Examples which are not in any way intended to limit the scope of the invention as claimed. The attached Figures are meant to be considered as integral parts of the specification and description of the invention. All references cited are herein specifically incorporated by reference for all that is described therein. The following examples are offered to illustrate, but not to limit the claimed invention.
  • In the experimental disclosure which follows, the following abbreviations apply PI (proteinase inhibitor), BBI (Bowman-Birk inhibitor), STI (Soybean Trypsin inhibitor); VEGF and VegF (vascular endothelial growth factor); ppm (parts per million); M (molar); mM (millimolar); μM (micromolar); mM (nanomolar); mol (moles); mmol (millimoles); μmol (micromoles); nmol (nanomoles); gm (grams); mg (milligrams); μg (micrograms); pg (picograms); L (liters); ml and mL (milliliters); μl and μL (microliters); cm (centimeters); mm (millimeters); μm (micrometers); nm (nanometers); U (units); V (volts); MW (molecular weight); sec (seconds); min(s) (minute/minutes); h(s) and hr(s) (hour/hours); ° C. (degrees Centigrade); QS (quantity sufficient); ND (not done); NA (not applicable); rpm (revolutions per minute); H2O (water); dH2O (deionized water); (HCl (hydrochloric acid); aa (amino acid); bp (base pair); kb (kilobase pair); kD (kilodaltons); cDNA (copy or complimentary DNA); DNA (deoxyribonucleic acid); ssDNA (single stranded DNA); dsDNA (double stranded DNA); dNTP (deoxyribonucleotide triphosphate); RNA (ribonucleic acid); MgCl2 (magnesium chloride); NaCl (sodium chloride); w/v (weight to volume); v/v (volume to volume); g (gravity); OD (optical density); Dulbecco's phosphate buffered solution (DPBS); SOC (2% Bacto-Tryptone, 0.5% Bacto Yeast Extract, 10 mM NaCl, 2.5 mM KCl); Terrific Broth (TB; 12 g/l Bacto Tryptone, 24 μl glycerol, 2.31 g/l KH2PO4, and 12.54 g/l K2HPO4); OD280 (optical density at 280 nm); OD600 (optical density at 600 nm); A405 (absorbance at 405 nm); Vmax (the maximum initial velocity of an enzyme catalyzed reaction); PAGE (polyacrylamide gel electrophoresis); PBS (phosphate buffered saline [150 mM NaCl, 10 mM sodium phosphate buffer, pH 7.2]); PBST (PBS+0.25% TWEEN® 20); PEG (polyethylene glycol); PCR (polymerase chain reaction); RT-PCR (reverse transcription PCR); SDS (sodium dodecyl sulfate); Tris(tris(hydroxymethyl)aminomethane); HEPES (N-[2-Hydroxyethyl]piperazine-N-[2-ethanesulfonic acid]); HBS (HEPES buffered saline); SDS (sodium dodecylsulfate); bME, BME and βME (beta-mercaptoethanol or 2-mercaptoethanol); Tris-HCl (tris[Hydroxymethyl]aminomethane-hydrochloride); Tricine (N-[tris(hydroxymethyl)-methyl]-glycine); CHES (2-(N-cyclo-hexylamino) ethane-sulfonic acid); TAPS (3-{[tris-(hydroxymethyl)-methyl]-amino}-propanesulfonic acid); CAPS (3-(cyclo-hexylamino)-propane-sulfonic acid; DMSO (dimethyl sulfoxide); DTF (1,4-dithio-DL-threitol); Glut and GSH (reduced glutathione); GSSG (oxidized glutathione); TCEP (Tris[2-carboxyethyl]phosphine); Ci (Curies) mCi (milliCuries); μCi (microCuries); TLC (thin layer achromatography); Ts (tosyl); Bn (benzyl); Ph (phenyl); Ms (mesyl); Et (ethyl), Me (methyl); Taq (Thermus aquaticus DNA polymerase); Klenow (DNA polymerase I large (Klenow) fragment); rpm (revolutions per minute); EGTA (ethylene glycol-bis(β-aminoethyl ether) N,N,N′,N′-tetraacetic acid); EDTA (ethylenediaminetetracetic acid); bla (β-lactamase or ampicillin-resistance gene); GE Healthcare (GE Healthcare, Chalfont St. Giles, United Kingdom); DNA2.0 (DNA2.0, Menlo Park, Calif.); OXOID (Oxoid, Basingstoke, Hampshire, UK); Megazyme (Megazyme International Ireland Ltd., Bray Business Park, Bray, Co., Wicklow, Ireland); Corning (Corning Life Sciences, Corning, N.Y.); (NEN (NEN Life Science Products, Boston, Mass.); Pharma AS (Pharma AS, Oslo, Norway); Dynal (Dynal, Oslo, Norway); Bio-Synthesis (Bio-Synthesis, Lewisville, Tex.); ATCC (American Type Culture Collection, Rockville, Md.); Gibco/BRL (Gibco/BRL, Grand Island, N.Y.); Sigma (Sigma Chemical Co., St. Louis, Mo.); Pharmacia (Pharmacia Biotech, Pisacataway, N.J.); NCBI (National Center for Biotechnology Information); Applied Biosystems (Applied Biosystems, Foster City, Calif.); Clontech (CLONTECH Laboratories, Palo Alto, Calif.); Operon Technologies (Operon Technologies, Inc., Alameda, Calif.); MWG Biotech (MWG Biotech, High Point, N.C.); Oligos Etc (Oligos Etc. Inc, Wilsonville, Oreg.); Bachem (Bachem Bioscience, Inc., King of Prussia, Pa.); Difco (Difco Laboratories, Detroit, Mich.); Mediatech (Mediatech, Herndon, Va.; Santa Cruz (Santa Cruz Biotechnology, Inc., Santa Cruz, Calif.); BioVeris (BioVeris Corp., Gaithersburg, Md.); Oxoid (Oxoid Inc., Ogdensburg, N.Y.); Worthington (Worthington Biochemical Corp., Freehold, N.J.); GIBCO BRL or Gibco BRL (Life Technologies, Inc., Gaithersburg, Md.); Millipore (Millipore, Billerica, Mass.); Bio-Rad (Bio-Rad, Hercules, Calif.); Invitrogen (Invitrogen Corp., San Diego, Calif.); NEB (New England Biolabs, Beverly, Mass.); Sigma (Sigma Chemical Co., St. Louis, Mo.); Pierce (Pierce Biotechnology, Rockford, Ill.); Takara (Takara Bio Inc. Otsu, Japan); Roche (Hoffmann-La Roche, Basel, Switzerland); EM Science (EM Science, Gibbstown, N.J.); Qiagen (Qiagen, Inc., Valencia, Calif.); Biodesign (Biodesign Intl., Saco, Me.); Aptagen (Aptagen, Inc., Herndon, Va.); Molecular Devices (Molecular Devices, Corp., Sunnyvale, Calif.); R&D Systems (R&D Systems, Minneapolis, Minn.); Stratagene (Stratagene Cloning Systems, La Jolla, Calif.); Marsh (Marsh Biosciences, Rochester, N.Y.); Bio-Tek (Bio-Tek Instruments, Winooski, Vt.); (Biacore (Biacore, Inc., Piscataway, N.J.); PeproTech (PeproTech, Rocky Hill, N.J.); SynPep (SynPep, Dublin, Calif.); and Microsoft (Microsoft, Inc., Redmond, Wash.).
  • Example 1 Production of BCE103-BBI Fusion Proteins in B. subtilis
  • In this Example, experiments conducted to produce BCE103-BBI fusion proteins in B. subtilis are described. The DNA sequence of the synthetic gene (Operon Technologies) coding for the pro-BBI protein with a C-terminal hexa-histidine tag used in these experiments is:
  • (SEQ ID NO:10)
    AACCTGCGTCTGTCTAAGCTTGGCCTGCTTATGAAATCAGACCATCAGCA
    CAGCAATGACGATGAGAGCTCTAAACCCTGTTGCGATCAATGCGCATGTA
    CGAAATCAAATCCTCCACAGTGTCGGTGTTCCGATATGCGTCTGAATAGC
    TGTCATAGTGCATGCAAAAGCTGTATCTGCGCCCTGAGTTATCCAGCTCA
    ATGTTTTTGCGTCGACATCACGGACTTCTGCTATGAGCCATGTAAACCAA
    GCGAGGACGATAAAGAGAACCATCATCACCATCACCAT
  • The protein sequence of pro-BBI with a C-terminal hexa-histidine tagged coded for by the above synthetic gene is:
  • (SEQ ID NO:11)
    NLRLSKLGLLMKSDHQHSNDDESSKPCCDQCACTKSNPPQCRCSDMRLNS
    CHSACKSCICALSYPAQCFCVDITDFCYEPCKPSEDDKENHHHHHH
  • The portion of the DNA sequence of the synthetic gene that codes for the major mature form of BBI is:
  • (SEQ ID NO:12)
    GACGATGAGAGCTCTAAACCCTGTTGCGATCAATGCGCATGTACGAAATC
    AAATCCTCCACAGTGTCGGTGTTCCGATATGCGTCTGAATAGCTGTCATA
    GTGCATGCAAAAGCTGTATCTGCGCCCTGAGTTATCCAGCTCAATGTTTT
    TGCGTCGACATCACGGACTTCTGCTATGAGCCATGTAAACCAAGCGAGGA
    CGATAAAGAGAAC
  • The protein sequence of the major mature form of BBI coded by the above synthetic gene is:
  • (SEQ ID NO:13)
    DDESSKPCCDQCACTKSNPPQCRCSDMRLNSCHSACKSCICALSYPAQCF
    CVDITDFCYEPCKPSEDDKEN
  • The PCR primers used to amplify the BBI gene for fusion to the BCE103 cellulase expression cassette in the pJ103 vector were:
  • BBIfusion_FW:
    (SEQ ID NO:14)
    5′ CAGCACGGATCCAGACGATGAGAGCTCTAAACCC 3′
    BBIHindIII_RV:
    (SEQ ID NO:15)
    5′ CTGCAGAAGCTTAAAAATAAAAAAACGGATTTCCTTCA
    GGAAATCCGTCCTCTGTTAACTTTTAGTTCTCTTTATCGTCCTCGC
    3′
    BBIHIS-HindIII_RV:
    (SEQ ID NO:16)
    5′ CTGCAGAAGCTTAAAAATAAAAAAACGGATT
    TCCTCAGGAAATCCGTCCTCTGTTAACTTTTAATGGTGATGGTGATGATG
    GTTCTC
    3′
  • The sequence of the aprE-BCE103-BBI-HisTag expression cassette (EcoRI-HindIII) that was cloned into the pJM103 integration vector is provided in FIG. 1. A schematic plasmid map of the pJM103BBIHis expression vector is provided in FIG. 2.
  • The alkaline cellulase (BCE103) gene (See, van Soligen, U.S. Pat. No. 6,063,611, hereby incorporated by reference) fused to the B. subtilis aprE promoter and signal sequence, was cloned from pUCAPR103 (Shaw et al., J. Mol. Biol., 320: 303-309 [2002]) as an EcoRI-BamHI fragment (i.e., a fragment that carries the coding sequence of the BCE103 catalytic domain and first cellulose binding domain linker only) into pJM103 (Perego, “Integrational vectors for genetic manipulation in Bacillus subtilis” In, Bacillus subtilis and Other Gram-positive Bacteria: Biochemistry, Physiology, and Molecular Genetics, Sonenshein, Hoch, and Losick (eds), American Society for Microbiology, Washington D.C., pp. 615-624 [1993]). A gene encoding the soybean Bowman-Birk protease inhibitor (BBI) (Swiss-Prot Accession # P01055; See, Odani and Ikenaka, J. Biochem., 71: 839-848 [1972]) with a C-terminal hexa-histidine tag (His-Tag) was synthesized by Operon Technologies (See, DNA sequence above). The BBI gene was amplified by PCR with primers (all primers were synthesized by MWG Biotech, Oligos Etc., or Operon Technologies) that generated a 5′ BamHI site in the correct reading frame with the BCE103 gene, and at the 3′ end introduced a strong transcriptional terminator (LAT, from the Bacillus licheniformis α-amylase gene) after the end of the BBI gene with a 3′ HindIII site for cloning into the pJM103 vector.
  • PCR fragments with or without a C-terminal His-Tag were generated with the primers BBIfusion_FW (SEQ ID NO:14) and BBIHISHindIII_RV (SEQ ID NO:16), or BBIfusion_FW (SEQ ID NO:14) and BBI-HindIII_RV (SEQ ID NO:15), respectively, using the synthetic BBI gene as a template. Unless indicated otherwise, PCR reactions were typically performed on a thermocycler for 30 cycles with High Fidelity Platinum Taq polymerase (Invitrogen) according to the instructions of the supplier (with an annealing temperature of 55° C.). The PCR fragments were cloned as BamHI-HindIII fragments into pJM103 carrying the aprE-BCE103 expression cassette. The correct gene sequence was verified by DNA sequencing.
  • Thus, as shown in FIG. 1, the N-terminus of the mature coding region of the BBI gene (with or without the His-Tag) was fused in frame to the C-terminal coding region of the first CBD (cellulose binding domain) linker sequence coded by the BCE103 cellulase gene. Thereby, the two CBD's of BCE103 (Shaw et al., supra) are replaced by BBI in the final expression vectors, pJM103BBI or pJM103BBIhis (See, FIG. 2). The aprE promoter controls the expression of the BCE103-BBI gene fusions (See, Ferrari et al., J. Bact., 170: 289-295 [1988]; and Henner et al., J. Bact., 170: 296-300 [1988]).
  • Competent Bacillus subtilis cells, BG3934comK (degUHy32, oppA, ΔspoIIE3501, ΔaprE, ΔnprE, Δepr, ΔispA, Δbpr, amyE::xylRPxylAcomK-phleo), were transformed with the expression plasmids, pJM103BBI or pJM103BBIhis. The bacteria were made competent by the induction of the comK gene under control of a xylose inducible promoter (Hahn et al., Mol. Microbiol., 21: 763-775 [1996]). The transformants were selected on Luria Broth agar (LA) plates containing 5 μg/ml chloramphenicol. To increase the expression by gene amplification, colonies were streaked and grown several times on LA plates with 25 μg/ml chloramphenicol until the growth rate with the antibiotic was similar to growth rate in the absence of chloramphenicol. The BCE103-BBI fusion protein was produced by growth in shake flasks at 37° C. in TSB medium (Tryptone Soya Broth from OXOID, 30 g/L) or in MBD medium, a MOPS based defined medium. MBD medium was made essentially as described (Neidhardt et al., J. Bacteriol., 119: 736-747 [1974]), except NH4Cl2, FeSO4, and CaCl2 were left out of the base medium, 3 mM K2HPO4 was used, and the base medium was supplemented with 60 mM urea, 75 g/L glucose, and 1% soytone. Also, the micronutrients were made up as a 100× stock containing in one liter, 400 mg FeSO4.7H2O, 100 mg MnSO4.H2O, 100 mg ZnSO4.7H2O, 50 mg CuCl2.2H2O, 100 mg CoCl2.6H2O, 100 mg NaMoO4.2H2O, 100 mg Na2B4O7.10H2O, 10 ml of 1M CaCl2, and 10 ml of 0.5 M sodium citrate.
  • BCE103-BBI fusion protein could be easily visualized in samples from cell free supernatants (after 24 h of growth in TSB medium or 48 h in MBD medium) as the major protein band on SDS-PAGE gels (10% NuPAGE in MES buffer, run as described by the manufacturer, Invitrogen) running at ˜44 kDa by using standard protein stains (e.g. GelCode Blue Stain Reagent; Pierce). The identity of the BCE103-BBI fusion protein was verified by immunoblots of SDS-PAGE gels using the protocols supplied by the manufacturer (BM Chromogenic Western Blotting Kit; Roche Applied Science using an anti-HisTag antibody or an anti-BCE103 cellulase polyclonal antibody for detection).
  • To determine the BCE103 activity, cellulase degradation was assessed qualitatively on LA cellulase indicator plates (with 1% carboxymethylcellulose stained with 0.2% Congo Red, or with 0.5% azo-CM-cellulose, Megazyme), or quantitatively by a direct assay in Assay Buffer (100 mM Tris pH 8.6, 0.005% Tween-80) on the culture broth using a the synthetic substrate, 4-nitrophenyl β-D-cellobioside (Sigma), using methods known in the art (See e.g., van Tilbeurgh et al., Meth. Enzymol., 160: 45-59 [1988]).
  • Trypsin inhibitory assays were performed in Assay Buffer to determine the BBI activity. Specifically, a standard curve was generated by making eleven 1:1 serial dilutions (100 μL BBI+100 μL Assay Buffer) of a 2 μg/mL standard BBI solution. The BBI standard was purified from a 1 mg/ml Trypsin-Chymotrypsin Inhibitor (Sigma Cat. #T-9777) solution in 20 mM MES pH 6.0 using a hydrophobic interaction column (POROS HP2, Phenyl column, Applied Biosystems). The column was equilibrated with 20 mM MES pH 6.0, loaded with 5 mg of the inhibitor, washed with the equilibration buffer, and then the BBI was eluted with water. Unknown BBI samples to be tested in the inhibitory assay were diluted as necessary, so that two or more data points would fall within the standard curve (usually 1:10, 1:100, 1:200, 1:1000, 1:2000 sample dilutions were tested and then the dilutions fine tuned if necessary). Each diluted BBI standard or sample, 20 μL, was added to 80 μL of 50 ng/ml bovine pancreatic trypsin (Worthington) (made by diluting a stock 1 mg/mL trypsin solution into Assay Buffer). For convenience, the standards and samples were arrayed in 96 well microtiter plates. The reactions were mixed and incubated 15 min at 25° C. After the incubation, 100 μL of the 0.5 mg/ml trypsin substrate (diluted in Assay Buffer from a 100 mg/ml solution in DMSO), Suc-AAPR-pNA (succinyl-Ala-Ala-Pro-Arg-para-nitroanilide, Bachem), was added, mixed and the OD (A405) was monitored for 15 min, with 1 time point recorded every 12 sec using a Spectra Max 250 (Molecular Devices). The data points were used to determine the Vmax for each reaction. The standard curve was generated by plotting Vmax versus BBI concentration and was fitted to a four-parameter curve. All data fitting was done using software supplied by the manufacturer (Molecular Devices). The BBI concentration of the unknown samples was calculated from the standard curve. Alternatively, the BBI activity was measured using the same protocol but by determining bovine pancreatic chymotrypsin (Worthington) inhibition (chymotrypsin was used at the same concentration as trypsin and chymotrypsin activity was measured by adding 100 μL of a 0.4 mg/ml chymotrypsin substrate, succinyl-Ala-Ala-Pro-Phe-para-nitroanilide, Bachem).
  • Titers from shake flask runs (500 ml MBD medium in 2.8 L Fernbach 6 baffled flasks, 37° C., 225 rpm, harvested 60 h after of growth) typically ranged from 0.4-0.9 mg/ml BCE activity and 40-150 μg/ml BBI trypsin inhibitory activity. However, it is contemplated that titers likely could be improved further by optimizing the bacterial strain, culture medium and growth conditions (aeration, temperature, time of harvest, etc.).
  • In addition to the BCE103 fusion to wild-type BBI, fusion proteins to BBI variants and fusion proteins with various linkers between BCE103 and BBI were produced using the methods outlined above, as described in the following Examples. In addition, fusion proteins were also produced when the BBI was fused to the 2nd CBD linker (BCE-cbdD-BBI; See, Example 4) making it possible to use the 1st CBD to aid in the purification process.
  • Example 2 Production of Peptides Substituted into the BBI Reactive Site Loops as BCE103-BBI Fusion Proteins
  • In this Example, experiments conducted to produce peptides substituted into the BBI reactive site loops as BCE103-BBI fusion proteins are described. The primers, as well as other sequences used in the various steps of these experiments are provided below. The sequence of 12BBIck81 from the BCE103 fusion site (at the BamHI) to the end of the gene is provided in FIG. 3. The CK37281 peptide sequences (ACYNLYGWTC (SEQ ID NO:9) are inserted into both the trypsin and chymotrypsin inhibitory loops.
  • The primers used to introduce an EcoRI site in the BBI gene using QuikChange® site-directed mutagenesis (Stratagene) were:
  • BowBeco-F
    5′-GATATGCGTCTGAATTCCTGTCATAGTGCAT (SEQ ID NO:17)
    BowBeco-R
    5′-ATGCACTATGACAGGAATTCAGACGCATATC (SEQ ID NO:18)
  • The sequences of the DNA oligonucleotides that were annealed and cloned in the BBI gene (SacI-EcoRI) to replace the trypsin inhibitory loop with the VegF binding peptide CK37281 were:
  • 1BBck81+
    (SEQ ID NO:19)
    5′-CTAAACCCTGTTGCGATCAATGCGCATGTTAATTTGTATGGGTGGAC
    TTGTCGCTGCAGCGATATGCGTCTG
    1BBck81−
    (SEQ ID NO:20)
    5′-AATTCAGACGCATATCGCTGCAGCGACAAGTCCACCCATACAAATTA
    TAACATGCGCATTGATCGCAACAGGGTTTAGAGCT
  • The sequences of the DNA oligonucleotides that were annealed and cloned in the BBI gene (EcoRI-SalI) to replace the chymotrypsin inhibitory loop with the VegF binding peptide CK37281 were:
  • 2BBck81+
    (SEQ ID NO:21)
    5′-AATTCCTGTCATAGTGCCTGCAAAAGCTGCGCATGTTATAACCTGTA
    CGGGTGGACCTGTTTTTGCG
    2BBck81−
    (SEQ ID NO:22)
    5′-TCGACGCAAAAACAGGTCCACCCGTACAGGTTATAACATGCGCAGCT
    TTTGCAGGCACTATGACAGG
  • The DNA sequences of the oligonucleotide pairs used to make cassettes to introduce peptides into the trypsin (SacI and EcoRI restriction sites) or chymotypsin (EcoRI and SalI restriction sites) reactive site loops of the synthetic BBI gene are provided below. These peptide coding sequences were then moved into the p2JM103BBI expression vector as SacI-SalI fragments.
  • Comstatin (1st loop)
    (SEQ ID NO:23)
    CTAAACCCTGTTGCGATCAATGCGCATGTGTTGTTCAGGACTGGGGTCAC
    CACCGTTGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:24)
    AATTCAGACGCATATCGCTGCAGCGACAACGGTGGTGACCCCAGTCCTGA
    ACAACACATGCGCATTGATCGCAACAGGGTTTAGAGCT
    Comstatin (2nd loop)
    (SEQ ID NO:25)
    CAAAAGCTGTATCTGCGTTGTTCAGGACTGGGGTCACCACCGTTGTTTTT
    GCG
    and
    (SEQ ID NO:26)
    TCGACGCAAAAACAACGGTGGTGACCCCAGTCCTGAACAACGCAGATACA
    GCTTTTGCATG
    C2c (1st loop)
    (SEQ ID NO:27)
    CTAAACCCTGTTGCGATCAATGCAGCTGTGGTCGTAAAATCCCGATCCAG
    TGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:28)
    AATTCAGACGCATATCGCTGCAGCGACACTGGATCGGGATTTTACGACCA
    CAGCTGCATTGATCGCAACAGGGTTTAGAGCT
    C3c (1st loop)
    (SEQ ID NO:29)
    CTAAACCCTGTTGCGATCAATGCGGTTGTGCTCGTTCTAACCTGGACGAA
    TGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:30)
    AATTCAGACGCATATCGCTGCAGCGACATTCGTCCAGGTTAGAACGAGCA
    CAACCGCATTGATCGCAACAGGGTTTAGAGCT
    C4c (1st loop)
    (SEQ ID NO:31)
    CTAAACCCTGTTGCGATCAATGCGGTTGTCAGCGTGCTCTGCCGATCCTG
    TGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:32)
    AATTCAGACGCATATCGCTGCAGCGACACAGGATCGGCAGAGCACGCTGA
    CAACCGCATTGATCGCAACAGGGTTTAGAGCT
    C5c (1st loop)
    (SEQ ID NO:33)
    CTAAACCCTGTTGCGATCAATGCCAGTGTGGTCGTCTGCACATGAAAACC
    TGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:34)
    AATTCAGACGCATATCGCTGCAGCGACAGGTTTTCATGTGCAGACGACCA
    CACTGGCATTGATCGCAACAGGGTTTAGAGCT
    Xa1 (2nd loop)
    (SEQ ID NO:35)
    AATTCCTGTCATAGTGCCTGCAAAAGCTGTATCTGCGCCCGTAGTTTGCC
    AGCTCAATGTTTTTGCG
    and
    (SEQ ID NO:36)
    TCGACGCAAAAACATTGAGCTGGCAAACTACGGGCGCAGATACAGCTTTT
    GCAGGCACTATGACAGG
    hSCC1 (1st loop)
    (SEQ ID NO:37)
    CTAAACCCTGTTGCGATCAATGCAACTGTACGTACTCAACCCCTCCACAG
    TGTCGCTGCAGCGATATGCGTCTG
    and
    (SEQ ID NO:38)
    AATTCAGACGCATATCGCTGCAGCGACACTGTGGAGGGGTTGAGTACGTA
    CAGTTGCATTGATCGCAACAGGGTTTAGAGCT
  • The DNA sequences of oligonucleotide primer pairs used to introduce peptide sequences into the trypsin or chymotrypsin reactive site loops using a QuikChange® II XL site-directed mutagenesis kit (Stratagene) are provided below. The reactions were performed as outlined by the manufacturer and described in this Example. Twenty cycles were performed with extensions of 6 minutes at 68° C., denaturations of 50 s at 95° C., and annealings at 55° C. for 50 s. After the cycles, a final extension was performed at 68° C. for 20 minutes.
  • 1A (2nd loop)
    (SEQ ID NO:39)
    CTGTATCTGCAAACGCTCAAAATCTCGTGGCTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:40)
    CGCAAAAACAGCCACGAGATTTTGAGCGTTTGCAGATACAGCTTTTGCA
    TG
    2B (2nd loop)
    (SEQ ID NO:41)
    CTGTATCTGCTGGTATAATCAAATGACAACATGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:42)
    CGCAAAAACATGTTGTCATTTGATTATACCAGCAGATACAGCTTTTGCA
    TG
    4A (2nd loop)
    (SEQ ID NO:43)
    CTGTATCTGCCATCAACTTGGCCCGAATTCATGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:44)
    CGCAAAAACATGAATTCGGGCCAAGTTGATGGCAGATACAGCTTTTGCA
    TG
    5A (2nd loop)
    (SEQ ID NO:45)
    CTGTATCTGCCATCCGTGGGCACCGTATTCTTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:46)
    CGCAAAAACAAGAATACGGTGCCCACGGATGGCAGATACAGCTTTTGCA
    TG
    6-1A (2nd loop)
    (SEQ ID NO:47)
    CTGTATCTGCAATCTTCATTATCTTCAACAGTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:48)
    CGCAAAAACACTGTTGAAGATAATGAAGATTGCAGATACAGCTTTTGCA
    TG
    7A (2nd loop)
    (SEQ ID NO:49)
    CTGTATCTGCACACCGTCTTTATCGCCCGTGTTTTTGCGTCGACATCAC
    and
    (SEQ ID NO:50)
    CGCAAAAACACGGGCGATAAAGAGACGGTGTGCAGATACAGCTTTTGCA
    TG
    8B (2nd loop)
    (SEQ ID NO:51)
    CTGTATCTGCCTTACAGATCAATCTAAACCGTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:52)
    CGCAAAAACACGGTTTAGATTGATCTGTAAGGCAGATACAGCTTTTGCA
    TG
    9A (2nd loop)
    (SEQ ID NO:53)
    CTGTATCTGCGTTACAACATCAATGGGCATGTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:54)
    CGCAAAAACACATGCCCATTGATGTTGTAACGCAGATACAGCTTTTGCA
    TG
    10B (2nd loop)
    (SEQ ID NO:55)
    CTGTATCTGCCGCGCATCACCGTATGATTGGTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:56)
    CGCAAAAACACCAATCATACGGTGATGCGCGGCAGATACAGCTTTTGCA
    TG
    11-1A (2nd loop)
    (SEQ ID NO:57)
    CTGTATCTGCTCAACACAAAAAATTCCGCAATGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:58)
    CGCAAAAACATTGCGGAATTTTTTGTGTTGAGCAGATACAGCTTTTGCA
    TG
    12B (2nd loop)
    (SEQ ID NO:59)
    CTGTATCTGCACAATTTCGCTCTGCAACATGTTTTTGCGTCGACATCAC
    and
    (SEQ ID NO:60)
    CGCAAAAACATGTTGCAGAGCGAAATTGTGTGCAGATACAGCTTTTGCA
    TG
    13A (2nd loop)
    (SEQ ID NO:61)
    CTGTATCTGCCCGGATCATGTTCCGCATCTTTGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:62)
    CGCAAAAACAAAGATGCGGAACATGATCCGGGCAGATACAGCTTTTGCA
    TG
    15-1A (2nd loop)
    (SEQ ID NO:63)
    CTGTATCTGCTCAGGCTTTCCGCTTTCTACATGTTTTTGCGTCGACATC
    AC
    and
    (SEQ ID NO:64)
    CGCAAAAACATGTAGAAAGCGGAAAGCCTGAGCAGATACAGCTTTTGCA
    TG
    1A6 (1st loop)
    (SEQ ID NO:65)
    TCAATGCGCATGTGAAGAGATCTGGACTATGCTTTGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:66)
    CGGAACACCGGCAAAGCATAGTCCAGATCTCTTCACATGCGCATTGATCG
    CAACAGG
    1A6 (2nd loop)
    (SEQ ID NO:67)
    CAAAAGCTGTGCTTGTGAAGAGATCTGGACTATGCTTTGCTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:68)
    ACGCAAAAGCAAAGCATAGTCCAGATCTCTTCACAAGCACAGCTTTTGCA
    TGCACTATG
    1C2 (1st loop)
    (SEQ ID NO:69)
    TCAATGCGCATGTTGGGCCCTTACTGTCAAAACATGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:70)
    CGGAACACCGGCATGTTTTGACAGTAAGGGCCCAACATGCGCATTGATCG
    CAACAGG
    1C2 (2nd loop)
    (SEQ ID NO:71)
    CAAAAGCTGTGCTTGTTGGGCCCTTACTGTCAAAACATGCTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:72)
    ACGCAAAAGCATGTTTTGACAGTAAGGGCCCAACAAGCACAGCTTTTGCA
    TGCACTATG
    2E2 (1st loop)
    (SEQ ID NO:73)
    TCAATGCGCATGTCTTACAGTACTGTGGACTACATGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:74)
    CGGAACACCGGCATGTAGTCCACAGTACTGTAAGACATGCGCATTGATCG
    CAACAGG
    2E2 (2nd loop)
    (SEQ ID NO:75)
    CAAAAGCTGTGCTTGTCTTACAGTACTGTGGACTACATGCTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:76)
    ACGCAAAAGCATGTAGTCCACAGTACTGTAAGACAAGCACAGCTTTTGCA
    TGCACTAT
    2E5 (1st loop)
    (SEQ ID NO:77)
    TCAATGCGCATGTACTCTTTGGAACAGATCTCCTTGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:78)
    CGGAACACCGGCAAGGAGATCTGTTCCAAAGAGTACATGCGCATTGATCG
    CAACAGG
    2E5 (2nd loop)
    (SEQ ID NO:79)
    CAAAAGCTGTGCTTGTACTCTTTGGAATCGATCTCCTTGCTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:80)
    ACGCAAAAGCAAGGAGATCGATTCCAAAGAGTACAAGCACAGCTTTTGCA
    TGCACTATG
    FGFns (1st loop)
    (SEQ ID NO:81)
    TCAATGCGCATGTACAAACATCGATTCTACTCCTTGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:82)
    CGGAACACCGGCAAGGAGTAGAATCGATGTTTGTACATGCGCATTGATCG
    CAACAGG
    FGFns (2nd loop)
    (SEQ ID NO:83)
    CAAAAGCTGTGCTTGCACAAACATCGATTCTACTCCTTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:84)
    ACGCAAAAACAAGGAGTAGAATCGATGTTTGTGCAAGCACAGCTTTTGCA
    TGCAACTATG
    FGFkr (1st loop)
    (SEQ ID NO:85)
    TCAATGCGCATGTACAAAAATCGATCGTACTCCTTGCCGGTGTTCCGATA
    TGCGGTC
    and
    (SEQ ID NO:86)
    CGGAACACCGGCAAGGAGTACGATCGATTTTTGTACATGCGCATTGATCG
    CAACAGG
    FGFkr (2nd loop)
    (SEQ ID NO:87)
    CAAAAGCTGTGCTTGCACAAAAATCGATCGTACTCCTTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:88)
    ACGCAAAAACAAGGAGTACGATCGATTTTTGTGCAAGCACAGCTTTTGCA
    TGCACTATG
    FGFh1 (1st loop)
    (SEQ ID NO:89)
    TCAATGCGCATGTCACCTGCAGACAACTGAAACATGCCGGTGTTCCGATA
    TGCGGTC
    and
    (SEQ ID NO:90)
    CGGAACACCGGCATGTTTCAGTTGTCTGCAGGTGACATGCGCATTGATCG
    CAACAGG
    FGFh1 (2nd loop)
    (SEQ ID NO:91)
    CAAAAGCTGTGCTTGCCACCTGCAGACAACTGAAACATGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:92)
    ACGCAAAAACATGTTTCAGTTGTCTGCAGGTGGCAAGCACAGCTTTTGCA
    TGCACTATG
    FGFgy (1st loop)
    (SEQ ID NO:93)
    TCAATGCGCATGTGGCTACTTCATCCCATCGATTTGCCGGTGTTCCGATA
    TGCGTC
    and
    (SEQ ID NO:94)
    CGGAACACCGGCAAATCGATGGGATGAAGTAGCCACATGCGCATTGATCG
    CAACAGG
    FGFgy (2nd loop)
    (SEQ ID NO:95)
    CAAAAGCTGTGCTTGCGGCTACTTCATCCCATCGATTTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:96)
    ACGCAAAAACAAATCGATGGGATGAAGTAGCCGCAAGCACAGCTTTTGCA
    TGCACTATG
    MM005 (1st loop)
    (SEQ ID NO:97)
    TCAATGCGCATGTTTACGTATCCTTGCTAACAAATGCCCGGTGTTCCGAT
    ATGCGTC
    and
    (SEQ ID NO:98)
    CGGAACACCGGCATTTGTTAGCAAGGATACGTAAACATGCGCATTGATCG
    CAACAGG
    MM005 (2nd loop)
    (SEQ ID NO:99)
    CAAAAGCTGTGCTTGCTTACGTATCCTTGCTAACAAATGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:100)
    ACGCAAAAACATTTGTTAGCAAGGATACGTAAGCAAGCACAGCTTTTGCA
    TGCACTATG
    MM007 (1st loop)
    (SEQ ID NO:101)
    GCGATCAATGCGCCTGCAGAACTCAACCATATCCTTTATGTCGGTGTTCC
    GATATGCGTCT
    and
    (SEQ ID NO:102)
    GGAACACCGACATAAAGGATATGGTTGAGTTCTGCAGGCGCATTGATCGC
    AACAGGGTTT
    MM007 (2nd loop)
    (SEQ ID NO:103)
    CAAAAGCTGTGCCTGCAGAACACAACCTTACCCACTTTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:104)
    ACGCAAAAACAAAGTGGGTAAGGTTGTGTTCTGCAGGCACAGCTTTTGCA
    TGCACTATG
    MM009 (2nd loop)
    (SEQ ID NO:105)
    CAAAAGCTGTGCCTGCCTGTTAACACCTACTCTTAACTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:106)
    ACGCAAAAACAGTTAAGAGTAGGTGTTAACAGGCAGGCACAGCTTTTGCA
    TGCACTATG
    MM010 (1st loop)
    (SEQ ID NO:107)
    TCAATGCGCATGCGCTCTTCCAACTCATTCTAACTGTCGGTGTTCCGATA
    TGCGTCT
    and
    (SEQ ID NO:108)
    CGGAACACCGACAGTTAGAATGAGTTGGAAGAGCGCATGCGCATTGATCG
    CAACAGG
    MM010 (2nd loop)
    (SEQ ID NO:109)
    CAAAAGCTGTGCCTGCGCGCTTCCTACACACTCTAACTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:110)
    ACGCAAAAACAGTTAGAGTGTGTAGGAAGCGCGCAGGCACAGCTTTTGCA
    TGCACTATG
    MM017 (2nd loop)
    (SEQ ID NO:111)
    CAAAAGCTGTGCCTGCCCTTTAGGCCTTTGCCCACCTTGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:112)
    ACGCAAAAACAAGGTGGGCAAAGGCCTAAAGGGCAGGCACAGCTTTTGCA
    TGCACTATG
    FGFps1 (2nd loop)
    (SEQ ID NO:113)
    AAGCTGTATCTGCTGGAACATCGATTCTACACCTTGTTTTTGCGTCGACA
    TCACGG
    and
    (SEQ ID NO:114)
    ACGCAAAAACAAGGTGTAGAATCGATGTTCCAGCAGATACAGCTTTTGCA
    TGCACT
    FGFps2 (1st loop)
    (SEQ ID NO:115)
    GCGATCAATGCATCTGTACTTGGATTGACAGTACTCCTTGTCGGTGTTCC
    GATATGCGTC
    and
    (SEQ ID NO:116)
    GGAACACCGACAAGGAGTACTGTCAATCCAAGTACAGATGCATTGATCGC
    AACAGGGTTT
    FGFps2 (2nd loop)
    (SEQ ID NO:117)
    AAGCTGTATCTGCACATGGATCGATAGTACTCCTTGTTTTTGCGTCGACA
    TCACGG
    and
    (SEQ ID NO:118)
    ACGCAAAAACAAGGTGTAGAATCGATCCATGTGCAGATACAGCTTTTGCA
    TGCACT
    FGFpsB (2nd loop)
    (SEQ ID NO:119)
    AAGCTGTATCTGTACATGGATCGATTGGACACCTTGTTTTTGCGTCGACA
    TCACGG
    and
    (SEQ ID NO:120)
    ACGCAAAAACAAGGTGTCCAATCGATCCATGTACAGATACAGCTTTTGCA
    TGCACT
    1A8 (2nd loop)
    (SEQ ID NO:121)
    CAAAAGCTGCGCATGTGTTACTACAGATTGGATCGAATGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:122)
    ACGCAAAAACATTCGATCCAATCTGTAGTAACACATGCGCAGCTTTTGCA
    TGCACTATG
    1A12 (2nd loop)
    (SEQ ID NO:123)
    CAAAAGCTGTGCCTGCCCAACACTTGGACTCATATGTGTTTTTGCGTCGA
    CATCACGGAC
    and
    (SEQ ID NO:124)
    ACGCAAAAACACATATGAGTCCAAAGTGTTGGGCAGGCACAGCTTTTGCA
    TGCACTATGAC
    1E11 (2nd loop)
    (SEQ ID NO:125)
    CAAAAGCTGCGCATGTTACTACTCTCAATTCCACCAATGTTTTTGCGTCG
    ACATCACGG
    and
    (SEQ ID NO:126)
    ACGCAAAAACATTGGTGGAATTGAGAGTAGTAACATGCGCAGCTTTTGCA
    TGCACTATG
    TGFps1 (2nd loop)
    (SEQ ID NO:127)
    CAAAAGCTGTCTTTGTCCGGAAAACGATAACGTTTCTCCTTGTAATTGCG
    TCGACATCACGGACTTCTG
    and
    (SEQ ID NO:128)
    TGTCGACGCAATTACAAGGAGAAACGTTATCGTTTTCCGGACAAAGACAG
    CTTTTGCATGCACTATGAC
  • The DNA sequences of the oligonucleotide pair used to make the cassette to introduce the MM021 peptide into the chymotrypsin reactive site loops of the p2JM103-lnk2-BBI expression vector are provided below. The cassette was ligated into the SphI and SalI restriction sites in the vector.
  • MM021 (2nd loop)
    CAAAAGCTGTGCTTGTAAACACAACGTACGTCTTTTATGTTTTTGCG (SEQ ID NO:129)
    and
    TCGACGCAAAAACATAAAAGACGTACGTTGTGTTTACAAGCACAGCTTTTGCATG (SEQ ID NO:130)
  • Libraries made of cysteine constrained peptides are popular reagents (e.g. the commercially available PhD-C7C Phage Display Peptide Library Kit; NEB) for selecting peptides that bind to substrates of interest. BBI has two cysteine constrained reactive site loops that are structurally similar to the peptide loops displayed in various methods used to select peptide binders. So, once a cysteine constrained binding peptide has been selected, BBI is suitable for use as a scaffold to present the peptide in a binding reaction.
  • The VegF binding peptide CK37281 (See e.g., co-pending U.S. Provisional Patent Application Ser. No. 60/520,403, filed Nov. 13, 2003, incorporated herein by reference) was grafted into BBI by replacing the trypsin, chymotrypsin, or both reactive site loops, with the CK37281 peptide sequence (ACYNLYGWTC) (SEQ ID NO:9) by using DNA oligonucleotide cassettes. To facilitate the construction, an EcoRI site was introduced in the coding region of the BBI gene (custom synthesized by Operon Technologies; See, Example 1) between the trypsin and chymotrypsin reactive site loops by QuikChange® site-directed mutagenesis, using methods described by the manufacturer (Stratagene) using the primers BowBeco-F and BowBeco-R, shown above (0.5 μmol of each primer was used in the QuikChange® reaction; after an initial denaturation step of 97° C. for 3 minutes, 18 PCR cycles of 68° C. for 12 minutes, 95° C. for 30 seconds and 55° C. for one minute, followed by a final extension reaction for 15 minutes at 68° C.).
  • To replace the trypsin inhibitory peptide loop, two DNA oligonucleotides (1BBCK81+ and 1BBCk81−) were annealed and ligated into the SacI and EcoRI restriction sites. Likewise, to replace the chymotrypsin inhibitory peptide loop, EcoRI and SalI sites were used for insertion of a DNA cassette made by annealing the oligonucleotides (2BBck81+ an 2BBck81−). The CK37281 peptide was grafted into both loops by inserting the CK37281 peptide in the chymotrypsin loop (using the oligonucleotides (2BBck81+ an 2BBck81−) after the trypsin loop was first replaced by the CK37281 peptide. BBI with the CK37281 peptide in the trypsin loop (1BBIck81) was moved into the pJM103BBI expression vector as a SacI-SphI fragment. BBI with the CK37281 in the chymotrypsin loop (2BBIck81), or both loops (12BBIck81), was moved into pJM103BBI as SacI-SalI fragments. The correct sequences were verified by DNA sequencing (the sequence of 12BBIck81 gene is shown in FIG. 3). The resulting vectors, pJM103-1BBIck81, pJM103-2BBIck81, or pJM103-12BBIck81, were used to transform B. subtilis BG3934comK, and the production of the BCE fusion proteins was determined as in Example 1 above.
  • The fusion protein running at ˜44 kDa was detected by SDS-PAGE to be the major protein present in the cell free broth. Although in some cases, there was significant degradation (up to 50%) of the BBI moiety (especially after >48 h of growth in MBD medium), as observed by the presence of a prominent protein band running at ˜34 kDa corresponding to the BCE103 catalytic core. In these cases, the titers of the BCE103 cellulase were similar to that measured with fusions to the wild-type BBI (Example 1), but the activity of the BBI (trypsin inhibition with 2BBIck81, or chymotrypsin inhibition with 1BBIck81) was generally about two fold less.
  • To reduce the proteolytic degradation of BBI variants during growth (i.e. decrease the amount of BCE103 cellulase core present on SDS-PAGE gels in comparison to the fusion protein), a Bacillus subtilis strain with nine protease genes deleted, BG6006 (degUHy32, oppA, ΔspoIIE3501, ΔaprE, ΔnprE, Δepr, ΔispA, Δbpr, Δvpr ΔwprA, Δmpr-ybjF, ΔnprB, amyE::lRPxylAcomK-ermC), was used as an expression host, and the growth temperature (35° C.) and aeration (200 rpm) were reduced. With these changes, a major fusion protein band (˜44 kDa) was observed on SDS-PAGE gels with an insignificant band present at the molecular weight expected for the BCE catalytic core protein (˜34 kDa).
  • In addition to the CK37281 peptide, a number of other cysteine constrained peptides were produced when substituted into the trypsin and/or chymotrypsin reactive site loops of BBI fused to the C-terminus of the BCE103 cellulase. Specific examples included:
      • (1) Peptides designed or selected as complement antagonists, compstatin introduced into the 1st or 2nd reactive site loops (See, Sahu et al., J. Immunol., 157: 884-891, [1996]), C2c (1st loop), C3c (1st loop), C4c (1st loop) and C5c (1st loop); or peptides selected in a Factor B binding reaction 1B, 2B, 4A, 5A, 6-1A, 7A, 8B, 9A, 10B, 11-1A, 12B, 13A, and 15-1A (all in 2nd loop);
      • (2) Peptides designed to bind to the proteases Factor Xa or stratum corenum chymotrypsin, Xal (2nd loop) or hSCCl (1st loop), respectively;
      • (3) Peptides selected in FGF5 binding reactions 1A6 (1st or 2nd loop), 1C2 (1st or 2nd loop), 2E2 (1st or 2nd loop), 2E5 (1st, 2nd or both loops), FGFns (1st or 2nd loop), FGFkr (1st or 2nd loop), FGFhl (1st or 2nd loop), FGFgy (1st or 2nd loop), MM005 (1st or 2nd loop), MM007 (1st, 2nd or both loops), MM009 (2nd loop), MM010 (1st, 2nd or both loops), MM017 (2nd loop), FGFps1 (2nd loop), FGFps2 (1st, 2nd or both loops), and FGFpsB (2nd loop); and
      • (4) Peptides selected in TGFβ-1 binding reactions 1A8 (2nd loop), 1A12 (2nd loop), 1E11 (2nd loop), TGFps1 (2nd loop), and MM021 (2nd loop).
  • The oligonucleotides used to introduce these peptides into either the trypsin (1st loop) or chymotrypsin (2nd loop) reactive site loops, and methods used to graft these peptides into BBI, are provided above. In all cases, fusion proteins were produced as determined by SDS-PAGE gels. However, with some substituted peptides, the amount of intact fusion protein was increased by reducing the proteolytic degradation as described above for the CK37281 substituted peptide.
  • Example 3 Activation of BBI By Thiol Reducing/Oxidizing Agents
  • After growth, the activity of the BBI (by trysin or chymotrypsin inhibition) is typically some 5-20 times lower than what would be expected from the activity of the BCE103 cellulase measured in the cell free supernatants (the two molecules should be present at a 1:1 molar ratio in the fusion protein). An increase in the activity of BBI (measured by either trypsin or chymotrypsin inhibition) in the BCE103-BBI fusion protein can be routinely obtained by adding bME, typically concentrations of 1-4 mM added to the MBD growth medium about 14 h after inoculation. The trypsin or chymotrypsin inhibitory activity of BBI in the fusion protein is also lower than expected when binding peptides (e.g. VegF binding peptide CK37281) replace the chymotrypsin or trypsin reactive site loop, respectively. As with the wild-type BBI, the inhibitory activity can be increased by treatment with bME. Unexpectedly, other thiol reducing agents (e.g., cysteine, reduced glutathione, DL-dithiothreitol and Tris[2 boxyethyl]phosphine) had small or negligible effects on the activation of BBI during growth in these experiments. Also, additions of antioxidants (e.g., ascorbic acid or DL-α-tocopherol acetate) or other adjuvants to the growth medium (e.g., isoleucine, soybean oil, Tween-80), or growth at 30° C. did not significantly improve the BCE103:BBI activity ratio.
  • Specifically, to determine the BBI activation during growth, cultures of B. subtilis BG6006 transformed with p2JM103-E3-2BBIck81 (See, Example 4, below) were grown in 40 ml MBD medium in 250 ml shake flasks at 37° C. for 13 h. Then, the thiol reducing agents indicated on the graph in FIG. 4 were added and cell supernatants harvested after 62 h of growth. The reagents 2-mercaptoethanol (BME), cysteine (Cys), reduced glutathione (Glut), and DL-dithiothreitol (DTT) were added to the growth medium to the final concentrations indicated on the graph provided in FIG. 4. Concentrations of 5 mM βME can result in better BCE103:BBI activity ratios but typically result in an overall decrease in both BCE103 and BBI titers (see FIG. 4), at least partially due to the reduction in bacterial growth caused by the added reagent. Titers of BCE103 and 2BBIck81 were determined using the assays described in Example 1.
  • BBI activation was also achieved after partial purification of the fusion proteins (e.g. BCE-lnk2-2BBIck81, see Example 4 below) by Q-Sepharose ion exchange chromatography.
  • The fusion protein was purified from cell free broth obtained from shake flasks or fermentor runs. The broth was filtered, diluted five to ten fold in water and the pH adjusted to pH 7.5-8.0. The diluted sample was loaded onto a column packed with Q-Sepharose resin (GE Healthcare). The column was washed with 50 mM Tris pH 7.5 and then washed again in the same buffer containing 300 mM NaCl. The fusion protein was eluted in the same buffer with 700 mM NaCl.
  • To activate the BBI, the pooled fusion protein fractions were diluted ten fold in Assay Buffer then treated with 2 mM βME and 0.2 mM oxidized glutathione (GSSG) with constant mixing on a stir plate or rocker platform for about 24 h at room temperature. The BBI could generally be activated to about 70-100% of the expected trypsin inhibitory activity based on the measured concentration of the BCE103 cellulase. Although the activation method outlined above generally yielded the best results, in some cases, in order to maximize the activation of a given sample, screens were performed in 96-well plates to determine the optimal conditions. Initially, the typical conditions screened were the dilution in Assay Buffer (e.g., a 2-50 fold dilution series), βME concentration (e.g., series between 0.5-5 mM) and oxidized glutathione concentration (e.g. 0 mM then a series of 1/20 to 1/2 the βME concentration).
  • The activation of the fusion protein BCE-lnk2-2BBIck81 is shown in FIG. 5. In this specific example, the fusion protein from a Q-Sepharose purification was diluted 1:10 in Dulbecco's PBS (Mediatech) with 0.005% TWEEN®-80. Beta-mercaptoethanol was added to a final concentration of 3 mM and incubated overnight at room temperature on a rocker. The sample was further incubated at room temperature for about 60 h with vigorous stirring on a magnetic stir plate. The titers of the BCE103 and 2BBIck81 (before and after βME treatment) were determined by cellulase assays and trypsin inhibitory assays, respectively.
  • In some embodiments, such as for activating BBI or it variants in cell free broth from large volume fermentations, it is desirable to reduce the dilution and βME concentration in the activation reaction. This can be accomplished by using higher concentrations of buffer (500 mM Tris pH 8.6), or changing to zwitterionic buffers such as CHES (also CAPS, Tricine, TAPS, and other suitable zwitterionic buffers). For example, cell free broth (or fusion protein fractions purified by ion exchange chromatography) was diluted 1:1 in 375 mM CHES pH 8.6 with 0.005% TWEEN®-80 then activated with 1 mM βME and 10 mM Na2SO3 and incubated with stirring at room temperature for about 24 h. BBI or its variants, as BCE103 cellulase fusion proteins, were routinely activated by this method to 70-100% of the expected value (based on BCE103 cellulase activities).
  • Example 4 Release of Free BBI/Variants by Cleavage of the BCE103-BBI Fusion Proteins
  • This Example describes experiments developed to release free BBI or its variants by cleavage of the BCE103-BBI fusion proteins.
  • The sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of pJM103-BBI to generate potential cleavage sites during culture growth between the BCE103 catalytic domain and BBI are provided below.
  • BCEsubBBI (a subtilisin-type sensitive peptide
    sequence)
    (SEQ ID NO:131)
    GATCCAGGTGGAGCTGCTTTAGTTGACGATGAGAGCT
    and
    (SEQ ID NO:132)
    CTCATCGTCAACTAAAGCAGCTCCACCTG
    BCEcbdLBBI (a portion of the 1st CBD)
    (SEQ ID NO:133)
    GATCCAGGTGAACCTGACCCAACTCCTCCATCTGATCCTGGAGAATACCC
    AGCTTGGGACGATGAGAGCT
    and
    (SEQ ID NO:134)
    CTCATCGTCCCAAGCTGGGTATTCTCCAGGATCAGATGGAGGAGTTGGGT
    CAGGTTCACCTG
    BCEproBBI (the entire pro peptide of BBI)
    (SEQ ID NO:135)
    GATCCGGCGAACCTGCGTCTGTCTAAGCTTGGCCTGCTTATGAAATCAGA
    CCATCAGCACAGCAATGACGATGAGAGCT
    and
    (SEQ ID NO:136)
    CTCATCGTCATTGCTGTGCTGATGGTCTGATTTCATAAGCAGGCCAAGCT
    TAGACAGACGCAGGTTCGCCG
    BCEshortproBBI (a C-terminal portion of the pro
    peptide of BBI)
    (SEQ ID NO:137)
    GATCCAAAATCAGACCATCAGCACAGCAATGACGATGAGAGCT
    and
    (SEQ ID NO:138)
    CTCATCGTCATTGCTGTGCTGATGGTCTGATTTTG
  • The sequences of the DNA oligonucleotide pair that was annealed and ligated into the BamHI and SacI sites of p2JM103-BBI to fuse BBI to the 2nd CBD linker of BCE103 cellulase are provided below.
  • BCEcbdDBBI
    (SEQ ID NO:139)
    GATCCAGGAGAACCGGACCCAACGCCCCCAAGTGATCCAGGAGAGTATCC
    AGCATGGGATTCAAATCAAATTTACACAAATGAAATTGTGTATCATAACG
    GTCAGTTATGGCAAGCGAAATGGTGGACACAAAATCAAGAGCCAGGTGAC
    CCATACGGTCCGTGGGAACCACTCAAATCTGACCCAGATTCAGACGATGA
    GAGCT
    and
    (SEQ ID NO:140)
    CTCATCGTCTGAATCTGGGTCAGATTTGAGTGGTTCCCACGGACCGTATG
    GGTCACCTGGCTCTTGATTTTGTGTCCACCATTTCGCTTGCCATAACTGA
    CCGTTATGATACACAATTTCATTTGTGTAAATTTGATTTGAATCCCATGC
    TGGATACTCTCCTGGATCACTTGGGGGCGTTGGGTCCGGTTCTCCTG
  • The peptide sequences susceptible to acid cleavage between aspartic acid and proline residues are provided below.
  • Linker 1—WGDPHY (SEQ ID NO:141)(Lidell et al., J. Biol. Chem. 278: 13944-51 [2003])
  • Linker 2—DNNDPI (SEQ ID NO:142)(Segalas et al., FEBS Lett., 371: 171-175 [1995])
  • Linker 3—VVADPN (SEQ ID NO:143)(Kempennan et al., Appl. Env. Microbiol., 69: 1589-1597
  • Oligonucleotide primers used to introduce a BssHII site into pJM103BBI by QuikChange® site-directed mutagenesis are provided below.
  • BCEbss-F
    (SEQ ID NO:144)
    5′-TGGCGTTCAGCAACATGAGCGCGCAGGCTGATGATTA
    BCEbss-R
    (SEQ ID NO:145)
    5′-TAATCATCAGCCTGCGCGCTCATGTTGCTGAACGCCA
  • Sequences of the DNA oligonucleotides that were annealed as a cassette (SalI-HindIII) to introduce HindIII and XhoI sites after the stop codon of BBI, to introduce a PacI site after the LAT, and remove the original HindIII site are provided below.
  • BCEterm+
    (SEQ ID NO:146)
    5′-GACATCACGGACTTCTGCTATGAGCCATGTAAACCAAGCGAGGACGA
    TAAAGAGAACTAAAAGCTTAACTCGAGGTTAACAGAGGACGGATTTCCTG
    AAGGAAATCCGTTTTTTTATTTTTAATTAAG
    BCEterm−
    (SEQ ID NO:147)
    5′-AGCTCTTAATTAAAAATAAAAAAACGGATTTCCTTCAGGAAATCCGT
    CCTCTGTTAACCTCGAGTTAAGCTTTTAGTTCTCTTTATCGTCCTCGCTT
    GGTTTACATGGCTCATAGCAGAAGTCCGTGATG
  • PCR primers used to generate the acid labile linkers provided above (i.e., Linker 1, Linker 2, and Linker 3) inserted between the BCE103 catalytic domain and BBI are provided below.
  • BCE103coreBssHII_FW
    (SEQ ID NO:148)
    5′-CAGCAACATGAGCGCGCAGGCTG
    linkerWGDPHY_RV
    (SEQ ID NO:149)
    5′-ATCGTCTGGATCCGGATAGTGGGGGTCTCCCCAAGATGCTGATTCTC
    TTATTTTTTCCC
    linkerDNNDPI_RV
    (SEQ ID NO:150)
    5′-ATCGTCTGGATCCGGTATGGGATCATTGTTGTCAGATGCTGATTCTC
    TTATTTTTTCCC
    linkerVVADPN_RV
    (SEQ ID NO:151)
    5′-ATCGTCTGGATCCGGGTTGGGATCTGCAACTACAGATGCTGATTCTC
    TTATTTTTTCCC
  • PCR primers used to generate the acid labile linkers provided above (i.e., Linker 1, Linker 2, and Linker 3) inserted into the 1st CBD linker.
  • BCE103corePstI_FW
    (SEQ ID NO:152)
    GCATAAGGAT GAGTCATCTG CAGCG
    LplusWGDPHY_RV
    (SEQ ID NO:153)
    5′-ATCGTCTGGATCCGGATAGTGGGGGTCTCCCCACGGTTCTCCTGGAT
    CAGATGGCGG
    LplusDNNDPI_RV
    (SEQ ID NO:154)
    5′-ATCGTCTGGATCCGGTATGGGATCATTGTTGTCCGGTTCTCCTGGAT
    CAGATGGCGG
    LplusVVADPN_RV
    (SEQ ID NO:155)
    5′-ATCGTCTGGATCCGGGTTGGGATCTGCAACTACCGGTTCTCCTGGAT
    CAGATGGCGG
  • Protein sequence of the acid labile linkers inserted between the BCE103 catalytic domain and BBI are provided below. The acid labile linkers are shown in bold type and the sequences from the first CBD domain are underlined.
  • Linker 1
    BCE-WGDPHY-PDP-BBI (SEQ ID NO:156)
    Linker 2
    BCE-DNNDPI-PDP-BBI (SEQ ID NO:157)
    Linker 3
    BCE-VVADPN-PDP-BBI (SEQ ID NO:158)
    LinkerPlus 1
    BCE-IPPSDPTPPSDPGEP-WGDPHY-PDP-BBI (SEQ ID NO:159)
    LinkerPlus 2
    BCE-IPPSDPTPPSDPGEP-DNNDPI-PDP-BBI (SEQ ID NO:160)
    LinkerPlus 3
    BCE-IPPSDPTPPSDPGEP-VVADPN-PDP-BBI (SEQ ID NO:161)
  • The sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of pJM103-BBI to generate potential cleavage sites between the BCE103 catalytic domain and BBI during the purification process are provided below.
  • BCEentBBI (Enteropeptidase cleaveage site)
    (SEQ ID NO:162)
    GATCCAGGTGGAGACGACGATGACAAAGACGATGAGAGCT
    and
    (SEQ ID NO:163)
    CTCATCGTCTTTGTCATCGTCGTCTCCACCTG
    BCEgenen1BBI (Genenase I cleavage site)
    (SEQ ID NO:164)
    GATCCAGGTGCTGCTCATTACGACGATGAGAGCT
    and
    (SEQ ID NO:165)
    CTCATCGTCGTAATGAGCAGCACCTG
  • The sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of pJM103-lnk2-1BBIck81 to generate potential cleavage sites between the BCE103 catalytic domain and BBI during the purification process are provided below.
  • BCEfurinBBI (Furin/Blisterase cleavage site)
    (SEQ ID NO:166)
    GATCCACGTGCTAAAAGAGACGATGAGAGCT
    and
    (SEQ ID NO:167)
    CTCATCGTCTCTTTTAGCACGTG
    BCEgenen2BBI (Genenase I cleavage site)
    (SEQ ID NO:168)
    GATCCAGGCGCTGCACACTACAACGACGATGAGAGCT
    and
    (SEQ ID NO:169)
    CTCATCGTCGTTGTAGTGTGCAGCGCCTG
    BCEfleBBI (Mpr cleavage site)
    (SEQ ID NO:170)
    GATCCATTCCTTGAAGACGATGAGAGCT
    and
    (SEQ ID NO:171)
    CTCATCGTCTTCAAGGAATG
  • Sequences of the oligonucleotide primer pairs used to introduce the E and E3 linkers in Linker 2 by QuikChange site-directed mutagenensis (Stratagene) are provided below.
  • BCE-Elnk-BBI (Mpr cleavage site)
    CCCATACCGGAGCCAGACGATGAGAGCTC (SEQ ID NO:172)
    and
    CATCGTCTGGCTCCGGTATGGGATCATTGTTG (SEQ ID NO:173)
  • The protein sequence of the E3 linker between the BCE103 catalytic domain and BBI was DNNDPIPEPDDESFNMPIPEP (SEQ ID NO:174). In this sequence, the E Linker is underlined and the sequence generated by faulty recombination in E. coli is shown in bold type. Cleavage by Mpr (or V8 protease) can occur after any of the three glutamic acids present in the E3 Linker. Thus, the structure was BCE-(SEQ ID NO:174)-BBI.
  • The sequences of the DNA oligonucleotide pairs that were annealed and ligated into the BamHI and SacI sites of p2JM103-lnk2-2BBIck81 to generate potential Genenase I cleavage sites between the BCE103 catalytic domain and BBI are provided below.
  • BCEgenen3BBI
    GATCCAGGCGCTGCACACTACAAATCAGACCATCAGCACAGCAATGACGATGAGAGCT (SEQ ID NO:175)
    and
    CTCATCGTCATTGCTGTGCTGATGGTCTGATTTGTAGTGTGCAGCGCCTG (SEQ ID NO:176)
    BCEgenen4BBI
    GATCCAGGCGCTGCACACTACGTAGAATTTCAAGACGATGAGAGCT (SEQ ID NO:177)
    and
    CTCATCGTCTTGAAATTCTACGTAGTGTGCAGCGCCTG (SEQ ID NO:178)
  • The protein sequence of a Genenase I sensitive cleavage site (also acid and Mpr sensitive) inserted between the BCE103 catalytic domain and BBI was DNNDPIPDPGAAHYVEFQ (SEQ ID NO:179). The Genenase I site (Gen4 Linker) is in bold type (cleavage occurs between the tyrosine and valine) (NEB) and Linker 2 is underlined. Cleavage by Mpr can also occur after the glutamic acid that follows the valine in the Gen4 linker. The sequence used herein was BCE-SEQ ID NO:179)-BBI.
  • Cleavage sites in the BCE103-lnk2-2BBIck81 fusion protein are indicated below. The C-terminal seven amino acids of the BCE103 catalytic domain (underlined), linker 2 sequence (bold type), and 2BBIck81 sequences are shown. The acid/heat labile Asp-Pro bonds are indicated with solid headed arrows and the Mpr sensitive bonds after glutamic acids are indicated with line headed arrows.
  • (SEQ ID NO:180)
             
    Figure US20090111160A1-20090430-P00001
          
    Figure US20090111160A1-20090430-P00002
       
    Figure US20090111160A1-20090430-P00002
       
    Figure US20090111160A1-20090430-P00001
    . . . KIRESAS DNNDPIPDPDDESSKPCCDQCACTKSNPPQCRCSDMR
                                         
    Figure US20090111160A1-20090430-P00001
    LNSCHSACKSCACYNLYGWTCFCVDITDFCYEPCKPSEDDKEN
  • In order to isolate free BBI or its variants, the BBI moiety needs to be cleaved from the BCE103-BBI fusion protein. In some embodiments, this is accomplished during growth, by proteases intrinsically produced by B. subtilis. In some alternative embodiments, this cleavage occurs after growth, during the purification process (e.g. by acid/heat or proteolytic cleavage). Linkers potentially susceptible to cleavage during growth were designed (See, above, sub, cbdL, pro, shortpro, and cbdD) and cloned into the pJM103BBI or p2JM103BBI expression vectors as BamHI-SacI cassettes. The production of fusion protein versus BCE103 catalytic domain was analyzed on SDS-PAGE gels as described in Example 1.
  • Little cleavage of the fusion protein was observed for all these linkers except with the pro linker, which was nearly completely cleaved so that very little intact fusion protein was observed on gels, although there was a large band corresponding to the BCE103 catalytic core. Unfortunately, this cleavage during growth resulted in negligible BBI activity measured in cell free supernatants and no BBI band could be identified on SDS-PAGE gels. Although it is not intended that the present invention be limited to any particular mechanism or theory, it is possible that the BBI is particularly sensitive to proteolytic degradation in its inactive form. Thus, cleavage during the purification process after activation is generally preferred.
  • In some embodiments, the bonds between aspartic acid and proline residues are cleaved by heat treatment at acidic pH as known in the art (See e.g., Landon, Meth. Enzymol., 47: 145-149 [1977]). The 1st CBD linker in the BCE103 cellulase has three Asp-Pro dipeptide sequences (See, FIG. 1) with the potential to be cleaved by acid/heat treatment. However, cleavage by acid/heat treatment at these sites was found to be inefficient. Protein sequences that are especially labile to acid/heat have been described in the literature, three of such sequences are WGDPHY (SEQ ID NO:141), DNNDPI (SEQ ID NO:142), and VVADPN (SEQ ID NO:143)(i.e., Linkers 1, 2 and 3).
  • Before these acid labile linkers were introduced into the BCE103-BBI expression vector, pJM103BBI, a BssHII site was introduced by QuikChange® XL (Stratagene) mutagenesis (using the manufacturer's methods; and described in Example 2 above, except 8 minute extension and 1 minute denaturation steps were used) in the aprE signal sequence coding region using the oligonucleotide primers BCEbss-F and BCEbss-R (provided above). Then, HindIII and XhoI sites were inserted in front of the LAT terminator (after the BBI stop codon) and a PacI site was added after the terminator (the original HindIII site after the LAT terminator was removed) by inserting an oligonucleotide cassette (BCEterm+ and BCEterm−; provided above) into the SalI and the original HindIII sites. This new vector was called “p2JM103BBI.”
  • The acid labile linker fragments were generated by PCR, using forward primer BCE103coreBssHII_FW with each of the reverse primers, linker WGDPHY_R, linker DNNDPI_RV, or linkerVVADPN_RV (the sequences of which are all provided above) and p2JM103BBI as the template (see Example 1 for the PCR protocol). The PCR fragments of 970 bp were digested with BamHI and PstI, the 154 bp fragments encoding the acid linker fragments were isolated from an agarose gel after electrophoresis, and ligated into the p2JM103 vector digested with BamHI and PstI that had also been purified from a gel. The linker sequences in the final expression vectors, p2JM103lnk1-BBI, p2JM103lnk2-BBI and p2JM103lnk3-BBI, were verified by DNA sequencing.
  • Competent B. subtilis strain BG3934comK or BG6006 were transformed with the plasmids, colonies selected on 5 μg/ml chloramphenicol LA plates and amplified to 25 μg/ml chloramphenicol as described in Example 1.
  • Similarly, the acid labile linkers were inserted into the first CBD linker. Specifically, PCR fragments were generated using the forward primer BCE103corePstI_FW with the reverse primers LplusWGDPHY_RV, LplusDNNDPI_RV, or LplusVVADPN_RV (See above, for the sequences) with p2JM103BBI as a template. The PCR fragments of about 150 bp were digested with BamHI and PstI, purified and ligated to the p2JM103BBI vector digested with BamHI and PstI. The correct sequences were verified by DNA sequencing and the plasmids p2JM103pllnk1-BBI, p2JM103pllnk2-BBI and p2JM103pllnk3-BBI were used to transform B. subtilis strains as described above.
  • After growth in MBD medium, the fusion proteins were purified by ion exchange chromatography essentially as described above (See, Example 2). The fusion protein was cleaved by treatment at 55° C. for 16 h in 10% formic acid. The BCE103 catalytic domain precipitated during the acid treatment and was removed by centrifugation. The free BBI in the supernatant was dried overnight on a SpeedVac. The sample was suspended in 50 mM Tris pH 8 before loading on the SDS-PAGE gel. By analysis of the protein stained SDS-PAGE gels, it was observed that acid cleavage was much more efficient in the fusion proteins where Linker 2 was inserted between the BCE103 catalytic domain and BBI (BCE-DNNDPI-PDP-BBI). This linker was found to be cleaved in a couple of hours at 75° C. in 20 mM glycine pH 2.
  • In alternative embodiments, the fusion protein was cleaved by treatment with a protease during the purification process. Linkers were designed with cleavage sites for glutamic acid specific proteases (e.g., Mpr or V8 protease), Furin/blisterase, Genenase I, and Enteropeptidase (Enterokinase). These linkers were introduced as oligonucleotide cassettes (See above, for the sequences) between the BCE103 catalytic core and BBI in the expression vector using the BamHI and SacI sites (See, FIG. 1). In the coding region of the original expression vector (pJM103BBI), there is a glutamic acid residue in the 1st CBD domain and at the third residue in BBI (See, FIG. 1), which is contemplated to be susceptible to cleavage by glutamic acid specific proteases such as B. subtilis Mpr (BsMpr) or V8 protease. However, neither BsMpr nor V8 protease were found to cleave the BCE-BBI fusion protein very efficiently at these sites. Thus, it was necessary to design other linkers that were susceptible to cleavage by these proteases.
  • The six acid labile linkers described above were tested for cleavage by BsMpr. These fusion proteins were cleaved by treatment for 16 h with 16 μg of BsMpr at room temperature. After cleavage, the BCE103 catalytic domain was precipitated by the addition of 10% formic acid and removed by centrifugation. The free BBI in the supernatant was dried overnight on a SpeedVac. The sample was suspended in 50 mM Tris pH 8, before loading on the SDS-PAGE. Similar to the acid cleavage, the BCE-DNNDPI-PDP-BBI (Linker 2) fusion protein was much more efficiently cleaved by BsMpr than any of the other linkers. Therefore, BBI and its variants were found to be effectively released from the BCE-DNNDPI-PDP-BBI fusion protein either by acid/heat treatment or proteolytic digestion with a glutamic acid specific protease such as BsMpr. Several other linkers designed for cleaved by Mpr (e.g., E, E3 linker, and fle, provided above) were tested but none of them had any advantages over Linker 2 (the E3 linker was generated by faulty recombination in E. coli after transformation with the QuikChange® site-directed mutagensis reaction designed to construct the E linker). As shown above, there are two acid/heat labile cleavage sites in Linker 2 and three sites sensitive to cleavage by Mpr.
  • Linkers designed for cleavage by Furin or Blisterase (NEB) (BCEfurinBBI), or Enteropeptidase (Enterokinase, NEB) (BCEentBBI) were tested, but none of these sequences were cleaved efficiently by the appropriate protease. Four linkers were also designed (BCEgenen1BBI, BCEgenen2BBI, BCEgenen3BBI, and BCEgenen4BBI) and tested for cleavage by Genenase I (NEB). Efficient cleavage of the fusion protein was observed only with the Gen4 Linker (BCEgenen4BBI). BsMpr was also found to efficiently cleave the Gen4 linker.
  • After activation of the purified BCE-lnk2-2BBIck81 fusion protein, cleavage by BsMpr does not go to completion as judged by SDS-PAGE gels. However, it was discovered that complete cleavage after activation of BCE-BBI fusion proteins with Linker 2 (or the Gen4 linker) can be accomplished by using the Mpr protease isolated from Bacillus licheniformis (BlMpr). While it is not intended that the present invention be limited to any particular mechanism, cleavage after the third amino acid in mature BBI appeared to be more sensitive to BlMpr while cleavage after the sixth amino acid from the C-terminus of BBI is more sensitive to BsMpr cleavage.
  • In some embodiments, after cleavage, the BBI is purified away from the BCE103 catalytic domain by selective acid precipitation (pH 3 or lower) of the BCE103 catalytic domain as described above, ion exchange chromatography (See, Example 5), or by selective binding of BBI on an anhydrotrypsin-agarose (Sigma) column loaded in 50 mM Tris pH 8.0, washed with 50 mM Tris pH 8.0 with 150 mM NaCl, then eluting bound BBI with 50 mM glycine pH 2.2 with 300 mM NaCl).
  • Example 5 Binding of BBIck81 to VegF
  • In this Example, experiments conducted to assess the binding of BBIck81 to VegF are described. The BCE103-lnk2-2BBIck81 fusion protein was produced in B. subtilis as described in Example 2. The fusion protein was purified, and the BBI trypsin inhibitory activity was increased by treatment with βME and oxidized glutathione as described in Example 3. The fusion protein was cleaved by BsMpr protease (See, Example 4) and the free 2BBIck81 was purified from the BCE103 catalytic domain by ion exchange chromatography using a Q-Sepharose column.
  • Briefly, after cleavage, the pH of the cleaved sample was adjusted to 5.5, the sample was th loaded onto the column (equilibrated with 25 mM MES pH 5.5). The free 2BBIck81 was washed through the column using 25 mM sodium acetate pH 5.0 while the BCE103 catalytic core remained bound to the resin. The 2BBIck81 fraction was concentrated by ultrafiltration and analyzed using an electrochemiluminescence (ECL) based binding assay (BioVeris). The Anti-VegF antibody (Santa Cruz) and VegF (PeproTech) were labeled with the electrochemiluminescent dye and biotin, respectively, as described by the manufacturer (BioVeris). Al materials were in Dulbecco's PBS (Mediatech) supplemented with 0.1% TWEEN®-80. An initial dilution series of Anti-VegF antibody (125, 250 and 500 ng/ml) and VegF (100, 150, 200 and 250 ng/ml) were tested in the binding assay to determine the concentrations of each that would give a robust ECL signal.
  • For testing 2BBIck81 binding, 50 μL of 500 ng/ml ECL labeled Anti-VegF antibody, 50 μL of 250 ng/ml biotinylated VegF and 100 μL 2BBIck81 (series of 12.5, 15, 31.25, 62.5, 125, 250 or 500 ng/ml) were incubated at room temperature for 2 h with shaking. Then, 50 μL of 0.2 mg/ml streptavidin coated beads were added and the reaction was incubated at room temperature for 30 minutes. The ECL signal was measured using a BioVeris M8/384 Analyzer as described by the manufacturer (BioVeris). As shown in FIG. 6, the ECL signal decreased as increasing concentrations of 2BBIck81 displaced more of the labeled Anti-VegF antibody bound to VegF attached to the magnetic beads.
  • Thus, the CK37281 peptide when grafted onto the chymotrypsin inhibitory loop of BBI (2BBIck81) competed with the Anti-VegF antibody for binding to VegF at micromolar concentrations. In fact, 2BBIck81 competed for VegF binding better than the synthesized CK37281 peptide itself (See, FIG. 6). The CK37281 peptide inserted into the trypsin inhibitory loop, 1BBIck81, also competed with the Anti-VegF antibody in the BioVeris assay. Thus, BBI was found to be useful as a scaffold to present active binding peptides selected by various screening methods.
  • Example 6 Use of Alternative Fusion Partners for the Production of 2BBIck81
  • In this Example, experiments conducted to evaluate alternative fusion partners are described. The DNA sequence of the oligonucleotide primers used to amplify the dsbC gene (E. coli) from pET-40b(+) are provided below. These primers generate a BssHII site at the 5′ end and a BamHI at the 3′ end for cloning into p2JM103-Gen4-2BBIck81.
  • DsbCBBI-F
    (SEQ ID NO:181)
    AACATGAGCGCGCAGGCTGATGACGCGGCAATTCAACAAACGTTAG
    DsbCBBI-R
    (SEQ ID NO:182)
    TCGTCTGGATCCGGTATGGGATCATTGTTGTCACCAGAACCACTAGTTGA
    TCCTTTACCGCTGGTCATTTTTTGGTG
  • The DNA sequences of the oligonucleotides that were annealed together to make a cassette (Alw44I-BamHI) for fusing the P. mendocina cutinase gene to BBI with Linker 2, are provided below.
  • CutinaseBBI+
    (SEQ ID NO:183)
    TGCACTTCTCTGCTTTGGTCTGTTGAACGCAGAGGTCTTGACAACAATGA
    TCCTATTCCG
    CutinaseBBI−
    (SEQ ID NO:184)
    GATCCGGAATAGGATCATTGTTGTCAAGACCTCTGCGTTCAACAGACCAA
    AGCAGAGG
  • Because the BBI moiety has seven disulfide bonds, it is contemplated that higher titers of active BBI will be obtained using fusion proteins other than the BCE103 cellulase catalytic domain. For example, in some embodiments, compositions such as thiol-disulfide oxidoreductases and/or protein disulfide isomerases find use as fusion proteins to help produce correctly folded BBI moieties. In this embodiment, no additional activation step is needed under most circumstances. In additional embodiments, other proteins produced at high titers in B. subtilis also find use as fusion partners. For example, the thermostable protein disulfide isomerase from the fungus Humicola insolens (hiPDI) has been used as a fusion partner to produce the light chain of immunoglobulin G (2 disulfides) in Bacillus brevis (See, Kajino et al., Appl. Env. Microbiol., 66: 638-642 [2000]).
  • To determine whether hiPDI could be a better fusion partner than BCE103 for the production of BBI, this hiPDI gene was synthesized (DNA2.0) and cloned into the expression vector, p2JM103-lnk2-2BBIck81 (See, Example 4) as a BssHII-SacI fragment. In designing the synthetic gene, codons occurring with high frequency in highly expressed B. subtilis genes were selected except in cases where restriction sites were introduced or deleted. In the final construction, the N-terminus of the mature hiPDI gene was fused to the AprE signal sequence and the C-terminus was fused to a linker with an Enteropeptidase cleaveage site (Kajino et al., Appl. Env. Microbiol., 66: 638-642 [2000]), which in turn was fused to 2BBIck81 (See, FIG. 7). This expression vector, p2JM-PDI-EK-2BBIck81, was used to transform B. subtilis BG6006 and the production of the fusion protein was determined in MBD medium (as described in Example 1) with or without 2 mM βME added 14 h after inoculation.
  • As determined by SDS-PAGE gels, the production of the PDI-2BBIck81 fusion protein was typically somewhat less than the BCE-2BBck81 grown under identical conditions. The BBI titers (trypsin inhibition) measured from the PDI-2BBIck81 cell free supernatants were also typically less than the BCE-2BBIck81 fusion. As with fusions to BCE103, the measured activities of BBI when fused to PDI were higher when grown in 2 mM βME and the BBI activity was increased by the addition of βME to the cell free supernatants after growth when grown in βME free medium (as described in Example 3). Thus, the thiol-disulfide oxidoreductase activity of PDI does not seem to significantly improve the titers of active 2BBIck81 in the fusion protein or obviate the need for activation of the BBI molecule.
  • In order to increase the reduction potential of the fusion protein, which was contemplated to improve the BBI titers during growth, DsbC from Escherichia coli was used as a fusion partner for 2BBIck81. The dsbC gene was amplified by PCR using Herculase Enhanced DNA polymerase as described by the manufacturer (Stratagene) using DsbCBBI-F and DsbCBBI-R as primers (sequences shown above) and pET-40b(+) (Novagen) as a template. The isolated PCR fragment was cloned into the vector p2JM103-Gen4-2BBIck81 (See, Example 4) as a BssHII-BamHI fragment. The correct sequence of the fusion gene was verified by DNA sequencing. In this case, the titers of the DsbC-2BBIck81 fusion protein were significantly lower than the BCE-2BBIck81 fusion protein as judged on SDS-PAGE gels and the titers of the active 2BBIck81 measured by trypsin inhibition were much lower as well.
  • Other proteins that are produced at high titers in B. subtilis find use as fusion partners for the production of BBI. One such protein is the cutinase from Pseudomonas mendocina, which has been expressed at high titers utilizing the aprE promoter from B. subtilis (See e.g., U.S. Pat. No. 5,429,950, herein incorporated by reference). The aprE-cutinase gene fusion as an EcoRI-Alw44I fragment (from pAK-15) was ligated with an Alw44I-BamHI linker oligonucleotide cassette (See, sequence above) into the p2JM103-lnk2-2BBIck81 (See, Example 4) that had been cut with EcoRI and BamHI. This cutinase-linker2-2BBkk81 expression vector (See, FIG. 8 for the EcoRI-BamHI aprE-cutinase-linker2 sequence) was used to transform B. subtilis BG6006 cells and the fusion protein was produced in MBD medium as described previously for the other fusion proteins (See, Example 1). In this case, the cutinase-linker2-2BBIck81 fusion protein was not the major band observed on SDS-PAGE gels and the measured lipase titers (as measured using the methods provided in U.S. Pat. No. 5,429,950) and BBI titers were much less (ca. 20 fold) than found with the BCE-2BBIck81 fusion protein. Also, the BBI titers in the cutinase fusion protein were not improved significantly when 3 mM βME was added to the growth medium. Thus, the highest titers of active 2BBIck81 was consistently obtained by activation of the BCE-2BBIck81 fusion protein. Nonetheless, it is contemplated that various fusion partners will find use in the present invention.
  • All patents and publications mentioned in the specification are indicative of the levels of those skilled in the art to which the invention pertains. All patents and publications are herein incorporated by reference to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference.
  • Having described the preferred embodiments of the present invention, it will appear to those ordinarily skilled in the art that various modifications may be made to the disclosed embodiments, and that such modifications are intended to be within the scope of the present invention.
  • Those of skill in the art readily appreciate that the present invention is well adapted to carry out the objects and obtain the ends and advantages mentioned, as well as those inherent therein. The compositions and methods described herein are representative of preferred embodiments, are exemplary, and are not intended as limitations on the scope of the invention. It is readily apparent to one skilled in the art that varying substitutions and modifications may be made to the invention disclosed herein without departing from the scope and spirit of the invention.
  • The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein. The terms and expressions which have been employed are used as terms of description and not of limitation, and there is no intention that in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.
  • The invention has been described broadly and generically herein. Each of the narrower species and subgeneric groupings falling within the generic disclosure also form part of the invention. This includes the generic description of the invention with a proviso or negative limitation removing any subject matter from the genus, regardless of whether or not the excised material is specifically recited herein.

Claims (15)

1. A composition comprising a fusion protein, wherein said fusion protein comprises a Bowman Birk Inhibitor (BBI) comprising SEQ ID NO:13 wherein the trypsin and/or chymotrypsin loop of said SEQ ID NO:13 is replaced with a peptide of interest.
2. The composition of claim 1, wherein said fusion protein comprises an amino acid selected from the group consisting of SEQ ID NOS:2, 4, 6 and 8.
3. The composition of claim 2, wherein said fusion protein is encoded by a nucleotide sequence selected from the group consisting of SEQ ID NOS:1, 3, 5, and 7.
4-7. (canceled)
8. The composition of claim 1, wherein said peptide of interest comprises the amino acid sequence set forth in SEQ ID NO:9.
9. The composition of claim 1, wherein said fusion protein comprises the amino acid sequence set forth in SEQ ID NO:4.
10-25. (canceled)
26. A protease inhibitor composition comprising a Bowman Birk Inhibitor (BBI) comprising SEQ ID NO:13, wherein the trypsin or chymotrypsin loop of said SEQ ID NO:13 is replaced with the amino acid sequence of SEQ ID NO:9.
27-33. (canceled)
34. The composition of claim 1, wherein said fusion protein binds a vascular endothelial growth factor.
35. The composition of claim 1, wherein said fusion protein further comprises at least one catalytic domain and a cleavage site.
36. The composition of claim 35, wherein said catalytic domain is selected from the group consisting of cellulase, cutinase, and disulfide isomerase.
37. The composition of claim 1, wherein said fusion protein further comprises at least one linker sequence.
38. The composition of claim 37, wherein said, wherein said linker sequence is selected from the group consisting of SEQ ID NOS:141-143.
39. The composition of claim 1, wherein said SEQ ID NO:13 is truncated at the N- and/or C-terminus, with as many as seven N-terminal residues up to the first cysteine (Cys8) and/or as many as nine C-terminal residues up to the last cysteine (Cys62) removed.
US12/172,921 2003-11-06 2008-07-14 Compositions comprising bowman-birk protease inhibitors and variants thereof Active US7696173B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/172,921 US7696173B2 (en) 2003-11-06 2008-07-14 Compositions comprising bowman-birk protease inhibitors and variants thereof

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US51815403P 2003-11-06 2003-11-06
US52040303P 2003-11-13 2003-11-13
US53095403P 2003-12-19 2003-12-19
US53120703P 2003-12-19 2003-12-19
US53118903P 2003-12-19 2003-12-19
US10/984,514 US7413877B2 (en) 2003-11-06 2004-11-08 Bacterial expression of bowman-birk protease inhibitors and variants thereof
US12/172,921 US7696173B2 (en) 2003-11-06 2008-07-14 Compositions comprising bowman-birk protease inhibitors and variants thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/984,514 Continuation US7413877B2 (en) 2003-11-06 2004-11-08 Bacterial expression of bowman-birk protease inhibitors and variants thereof

Publications (2)

Publication Number Publication Date
US20090111160A1 true US20090111160A1 (en) 2009-04-30
US7696173B2 US7696173B2 (en) 2010-04-13

Family

ID=34596198

Family Applications (8)

Application Number Title Priority Date Filing Date
US10/581,142 Abandoned US20080113917A1 (en) 2003-11-06 2004-11-06 Tgfbeta Supported and Binding Peptides
US10/984,410 Expired - Fee Related US7485618B2 (en) 2003-11-06 2004-11-08 FGF-5 supported and binding peptides
US10/984,514 Active 2025-05-12 US7413877B2 (en) 2003-11-06 2004-11-08 Bacterial expression of bowman-birk protease inhibitors and variants thereof
US10/984,270 Expired - Fee Related US7524816B2 (en) 2003-11-06 2004-11-08 Peptides and supported peptides for treating skin diseases
US12/033,848 Abandoned US20080269139A1 (en) 2003-11-06 2008-02-19 FGF-5 Supported and Binding Peptides
US12/172,921 Active US7696173B2 (en) 2003-11-06 2008-07-14 Compositions comprising bowman-birk protease inhibitors and variants thereof
US12/403,272 Abandoned US20110230405A1 (en) 2003-11-06 2009-03-12 Peptides and Supported Peptides for Treating Skin Diseases
US13/609,116 Abandoned US20130065829A1 (en) 2003-11-06 2012-09-10 Peptides and supported peptides for treating skin diseases

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US10/581,142 Abandoned US20080113917A1 (en) 2003-11-06 2004-11-06 Tgfbeta Supported and Binding Peptides
US10/984,410 Expired - Fee Related US7485618B2 (en) 2003-11-06 2004-11-08 FGF-5 supported and binding peptides
US10/984,514 Active 2025-05-12 US7413877B2 (en) 2003-11-06 2004-11-08 Bacterial expression of bowman-birk protease inhibitors and variants thereof
US10/984,270 Expired - Fee Related US7524816B2 (en) 2003-11-06 2004-11-08 Peptides and supported peptides for treating skin diseases
US12/033,848 Abandoned US20080269139A1 (en) 2003-11-06 2008-02-19 FGF-5 Supported and Binding Peptides

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/403,272 Abandoned US20110230405A1 (en) 2003-11-06 2009-03-12 Peptides and Supported Peptides for Treating Skin Diseases
US13/609,116 Abandoned US20130065829A1 (en) 2003-11-06 2012-09-10 Peptides and supported peptides for treating skin diseases

Country Status (9)

Country Link
US (8) US20080113917A1 (en)
EP (4) EP1680507B1 (en)
JP (2) JP4843497B2 (en)
AT (4) ATE530560T1 (en)
CA (1) CA2544820C (en)
DE (2) DE602004029393D1 (en)
DK (2) DK1692158T3 (en)
ES (1) ES2375360T3 (en)
WO (4) WO2005046710A2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9486513B1 (en) 2010-02-09 2016-11-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US9657085B1 (en) 2009-02-09 2017-05-23 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US9878023B1 (en) 2010-02-09 2018-01-30 David Gordon Bermudes Protease inhibitor: protease sensitive expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US10087451B2 (en) 2006-09-22 2018-10-02 Aviex Technologies Llc Live bacterial vectors for prophylaxis or treatment
US10857233B1 (en) 2010-02-09 2020-12-08 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria

Families Citing this family (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE555119T1 (en) * 2003-11-06 2012-05-15 Danisco Us Inc EXPRESSION OF PROTEASE INHIBITORS AND VARIANTS THEREOF IN FILAMENTOUS FUNGI
EP1680507B1 (en) * 2003-11-06 2011-06-22 Genencor International, Inc. Bacterial expression of protease inhibitors and variants thereof
WO2006044650A2 (en) * 2004-10-15 2006-04-27 Genencor International, Inc. Competitive differential screening
US9084734B2 (en) * 2005-05-05 2015-07-21 Danisco Us Inc. Peptide personal care compositions and methods for their use
CA2610976C (en) 2005-06-10 2020-11-24 Robbie Woodman Use of stefin a as a scaffold protein
US8603782B2 (en) * 2005-07-13 2013-12-10 Amgen Inc. Heat lysis production of proteins
GB0601684D0 (en) * 2006-01-27 2006-03-08 Reckitt Benckiser Uk Ltd Composition, process for preparation and method of use
WO2007092465A2 (en) * 2006-02-07 2007-08-16 Dyao Pharmaceutical Corporation Topical delivery compositions
JP5500989B2 (en) * 2006-09-16 2014-05-21 ソイ ラブズ エルエルシー Products and methods using soy peptides to reduce total cholesterol and LDL cholesterol levels
US20080090745A1 (en) * 2006-10-13 2008-04-17 Fox Bryan P Expression of Streptomyces subtilism inhibitor (SSI) proteins in Bacillus and Streptomyces sp.
BRPI0721285A2 (en) 2006-12-18 2014-03-25 Danisco Us Inc Genencor Div LACASE MEDIATORS AND METHODS OF USE
JP2010512787A (en) 2006-12-21 2010-04-30 ダニスコ・ユーエス・インク、ジェネンコー・ディビジョン Composition and use of Bacillus sp. 195 alpha-amylase polypeptide.
WO2008153925A2 (en) * 2007-06-06 2008-12-18 Danisco Us, Inc., Genencor Division Methods for improving multiple protein properties
US20090048136A1 (en) * 2007-08-15 2009-02-19 Mcdonald Hugh C Kappa-carrageenase and kappa-carrageenase-containing compositions
EP2229439A1 (en) * 2007-10-31 2010-09-22 Danisco US Inc. Use and production of neutral metallproteases in a serine protease-free background
MX2010004325A (en) * 2007-11-01 2010-05-20 Danisco Us Inc Signal sequences and co-expressed chaperones for improving protein production in a host cell.
GB0722332D0 (en) * 2007-11-14 2007-12-27 Reckitt Benckiser Uk Ltd Personal care article
AU2008349567A1 (en) * 2008-01-29 2009-08-06 Danisco Us Inc. Expression of streptomyces subtilisin inhibitor (SSI) proteins in bacillus and streptomyces sp.
RU2526516C2 (en) 2008-02-04 2014-08-20 ДАНИСКО ЮЭс ИНК. Ts23 alpha-amylase versions with altered properties
US7772181B2 (en) * 2008-10-15 2010-08-10 Danisco Us Inc. Personal care compositions comprising modified variant Bowman Birk Protease Inhibitors
US7803902B2 (en) 2008-10-15 2010-09-28 Danisco Us Inc. Modified variant bowman birk protease inhibitors
EP2344531B1 (en) * 2008-10-15 2014-12-10 Danisco US Inc. Modified variant bowman birk protease inhibitors
EP2406373B1 (en) 2009-03-10 2014-05-28 Danisco US Inc. Bacillus megaterium strain dsm90-related alpha-amylases, and methods of use, thereof
US20100247693A1 (en) * 2009-03-24 2010-09-30 Marini Jan L Cosmetic formulation to treat rosacea telangiectasia
CN102388131B (en) 2009-04-08 2014-04-30 丹尼斯科美国公司 Halomonas strain WDG195-related alpha-amylases, and methods of use thereof
CN105238801A (en) 2009-04-24 2016-01-13 丹尼斯科美国公司 Proteases with modified pro regions
WO2010127231A2 (en) * 2009-05-01 2010-11-04 Signal Investment And Management Co. Moisturizing antimicrobial composition
AR076941A1 (en) 2009-06-11 2011-07-20 Danisco Us Inc BACILLUS CEPA FOR A GREATER PROTEIN PRODUCTION
US20110052676A1 (en) * 2009-09-01 2011-03-03 James Vincent Gruber Composition For Delaying Cellular Senescence
US8445445B2 (en) 2009-09-15 2013-05-21 Five Prime Therapeutics, Inc. Method of promoting hair growth using FGFR4 extracellular domains
GB0921001D0 (en) * 2009-11-30 2010-01-13 Aqua Bio Technology Asa Products and uses
MX356282B (en) 2009-12-09 2018-05-22 Danisco Us Inc Compositions and methods comprising protease variants.
EP2512501A4 (en) 2009-12-17 2014-01-01 Five Prime Therapeutics Inc Hair growth methods using fgfr3 extracellular domains
EP2785730A1 (en) * 2009-12-30 2014-10-08 Solae, LLC Method for recovering kunitz-trypsin inhibitor proteins from a soy processing stream
US9687591B2 (en) 2010-03-31 2017-06-27 Agency For Science, Technology And Research Building stratified biomimetic tissues and organs using crosslinked ultrashort peptide hydrogel membranes
US8999916B2 (en) 2010-03-31 2015-04-07 Agency For Science, Technology And Research Crosslinked peptide hydrogels
ES2574056T3 (en) 2010-03-31 2016-06-14 Agency For Science, Technology And Research Linear amphiphilic peptide / peptoid and hydrogel comprising the same
AR080886A1 (en) 2010-04-15 2012-05-16 Danisco Us Inc COMPOSITIONS AND METHODS THAT INCLUDE VARIABLE PROTEASES
DK3095861T3 (en) 2010-05-06 2019-09-23 Procter & Gamble CONSUMER PRODUCTS WITH PROTEASE VARIETIES
US8481038B2 (en) 2010-11-15 2013-07-09 Five Prime Therapeutics, Inc. Treatment of cancer with elevated dosages of soluble FGFR1 fusion proteins
EP4230735A1 (en) 2011-05-05 2023-08-23 Danisco US Inc. Compositions and methods comprising serine protease variants
WO2012151480A2 (en) 2011-05-05 2012-11-08 The Procter & Gamble Company Compositions and methods comprising serine protease variants
AR087745A1 (en) 2011-08-31 2014-04-16 Danisco Us Inc COMPOSITIONS AND METHODS THAT INCLUDE A VARIANT OF LIPOLITIC ENZYME
CN104024407A (en) 2011-12-22 2014-09-03 丹尼斯科美国公司 Compositions and methods comprising lipolytic enzyme variant
US9120841B2 (en) 2012-09-28 2015-09-01 Agency For Science, Technology And Research Amphiphilic linear peptidepeptoid and hydrogel comprising the same
KR20150067336A (en) 2012-10-12 2015-06-17 다니스코 유에스 인크. Compositions and methods comprising a lipolytic enzyme variant
CA2889864C (en) 2012-11-05 2023-02-28 Danisco Us Inc. Compositions and methods comprising thermolysin protease variants
EP2935573A1 (en) 2012-12-19 2015-10-28 Danisco US Inc. Novel mannanase, compositions and methods of use thereof
EP3110833B1 (en) 2013-05-29 2020-01-08 Danisco US Inc. Novel metalloproteases
JP6367930B2 (en) 2013-05-29 2018-08-01 ダニスコ・ユーエス・インク Novel metalloprotease
EP3882346A1 (en) 2013-05-29 2021-09-22 Danisco US Inc. Novel metalloproteases
US20160160202A1 (en) 2013-05-29 2016-06-09 Danisco Us Inc. Novel metalloproteases
ES2797483T3 (en) 2013-07-19 2020-12-02 Danisco Us Inc Compositions and Methods Comprising a Lipolytic Enzyme Variant
WO2015038792A1 (en) 2013-09-12 2015-03-19 Danisco Us Inc. Compositions and methods comprising lg12-clade protease variants
EP3055328A1 (en) * 2013-10-11 2016-08-17 F. Hoffmann-La Roche AG Nsp4 inhibitors and methods of use
JP2016535770A (en) 2013-11-01 2016-11-17 スフェリウム バイオメッド エス.エル. Inclusion body for transdermal delivery of therapeutic and cosmetic substances
US20160312204A1 (en) 2013-12-13 2016-10-27 Danisco Us Inc. Serine proteases of bacillus species
US10533165B2 (en) 2013-12-13 2020-01-14 Danisco Us Inc Serine proteases of the bacillus gibsonii-clade
US9737592B1 (en) * 2014-02-14 2017-08-22 David Gordon Bermudes Topical and orally administered protease inhibitors and bacterial vectors for the treatment of disorders and methods of treatment
EP3119884B1 (en) 2014-03-21 2019-07-10 Danisco US Inc. Serine proteases of bacillus species
EP3207129B1 (en) 2014-10-17 2019-11-20 Danisco US Inc. Serine proteases of bacillus species
EP3212783A1 (en) 2014-10-27 2017-09-06 Danisco US Inc. Serine proteases
DK3212662T3 (en) 2014-10-27 2020-07-20 Danisco Us Inc serine proteases
US20180010074A1 (en) 2014-10-27 2018-01-11 Danisco Us Inc. Serine proteases of bacillus species
EP3212782B1 (en) 2014-10-27 2019-04-17 Danisco US Inc. Serine proteases
US20170335306A1 (en) 2014-10-27 2017-11-23 Danisco Us Inc. Serine proteases
US20180216090A1 (en) 2015-03-12 2018-08-02 Danisco Us Inc. Compositions and methods comprising lg12-clade protease variants
EP3872174B1 (en) 2015-05-13 2023-03-01 Danisco US Inc. Aprl-clade protease variants and uses thereof
CN107922934A (en) 2015-06-17 2018-04-17 丹尼斯科美国公司 Protease with modified pre-peptide region
US11499146B2 (en) 2015-06-17 2022-11-15 Danisco Us Inc. Bacillus gibsonii-clade serine proteases
CN107022432A (en) * 2016-01-29 2017-08-08 高露洁-棕榄公司 Cleasing compositions
EP3845642B1 (en) 2016-05-05 2023-08-09 Danisco US Inc. Protease variants and uses thereof
JP2019523645A (en) 2016-05-31 2019-08-29 ダニスコ・ユーエス・インク Protease variants and uses thereof
CA3027745A1 (en) 2016-06-17 2017-12-21 Danisco Us Inc. Protease variants and uses thereof
EP4212622A3 (en) 2016-12-21 2023-11-29 Danisco US Inc. Bacillus gibsonii-clade serine proteases
EP3559227A1 (en) 2016-12-21 2019-10-30 Danisco US Inc. Protease variants and uses thereof
US11298391B1 (en) * 2017-04-27 2022-04-12 Lorenol Laboratories, Inc. Topical skin health improvement compositions and administrations thereof
US20200354708A1 (en) 2017-11-29 2020-11-12 Danisco Us Inc. Subtilisin variants having improved stability
US20210214703A1 (en) 2018-06-19 2021-07-15 Danisco Us Inc Subtilisin variants
US20210363470A1 (en) 2018-06-19 2021-11-25 Danisco Us Inc Subtilisin variants
WO2020046613A1 (en) 2018-08-30 2020-03-05 Danisco Us Inc Compositions comprising a lipolytic enzyme variant and methods of use thereof
EP3875591A4 (en) * 2018-10-31 2022-09-21 Qingdao Vland Biotech Group Co. Ltd. Method for producing washing enzyme having protease resistance
EP3887515A1 (en) 2018-11-28 2021-10-06 Danisco US Inc. Subtilisin variants having improved stability
WO2020242858A1 (en) 2019-05-24 2020-12-03 Danisco Us Inc Subtilisin variants and methods of use
CN112062828B (en) * 2019-06-10 2022-04-29 温州医科大学 FGF-5 polypeptide inhibitor and hair growth promoting application thereof
WO2021146255A1 (en) 2020-01-13 2021-07-22 Danisco Us Inc Compositions comprising a lipolytic enzyme variant and methods of use thereof
US20240034960A1 (en) 2020-08-27 2024-02-01 Danisco Us Inc Enzymes and enzyme compositions for cleaning
CN113307857B (en) * 2021-01-14 2022-11-01 艾时斌 Scaffold proteins derived from epidermal growth factor, lectin and Tat proteins
WO2023278297A1 (en) 2021-06-30 2023-01-05 Danisco Us Inc Variant lipases and uses thereof
WO2023034486A2 (en) 2021-09-03 2023-03-09 Danisco Us Inc. Laundry compositions for cleaning
US20230365897A1 (en) 2021-12-16 2023-11-16 The Procter & Gamble Company Fabric and home care composition including a protease
WO2023114932A2 (en) 2021-12-16 2023-06-22 Danisco Us Inc. Subtilisin variants and methods of use
WO2023114939A2 (en) 2021-12-16 2023-06-22 Danisco Us Inc. Subtilisin variants and methods of use
WO2023114795A1 (en) 2021-12-16 2023-06-22 The Procter & Gamble Company Automatic dishwashing composition comprising a protease
WO2023114936A2 (en) 2021-12-16 2023-06-22 Danisco Us Inc. Subtilisin variants and methods of use
WO2023168234A1 (en) 2022-03-01 2023-09-07 Danisco Us Inc. Enzymes and enzyme compositions for cleaning
WO2024050343A1 (en) 2022-09-02 2024-03-07 Danisco Us Inc. Subtilisin variants and methods related thereto
WO2024050346A1 (en) 2022-09-02 2024-03-07 Danisco Us Inc. Detergent compositions and methods related thereto

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5411873A (en) * 1984-05-29 1995-05-02 Genencor, Inc. Process for producing heterologous polypeptides
US5429950A (en) * 1992-03-30 1995-07-04 Genencor International, Inc. Heterologous gene expression in bacillus subtilis: fusion approach
US5679543A (en) * 1985-08-29 1997-10-21 Genencor International, Inc. DNA sequences, vectors and fusion polypeptides to increase secretion of desired polypeptides from filamentous fungi
US6063611A (en) * 1996-03-12 2000-05-16 Genencor International, Inc. Alkaline cellulase and method of producing same
US6537968B1 (en) * 2000-07-24 2003-03-25 Alphamed Pharmaceuticals Corp Treatment of lupus erythematosus
US6872563B1 (en) * 1999-10-05 2005-03-29 President And Fellows Of Harvard College Compositions and methods for production of disulfide bond containing proteins in host cells

Family Cites Families (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3755560A (en) * 1971-06-30 1973-08-28 Dow Chemical Co Nongreasy cosmetic lotions
DE2437090A1 (en) * 1974-08-01 1976-02-19 Hoechst Ag CLEANING SUPPLIES
JPS51125468A (en) 1975-03-27 1976-11-01 Sanyo Chem Ind Ltd Method of preparing resins of high water absorbency
US4152416A (en) * 1976-09-17 1979-05-01 Marra Dorothea C Aerosol antiperspirant compositions delivering astringent salt with low mistiness and dustiness
US4421769A (en) * 1981-09-29 1983-12-20 The Procter & Gamble Company Skin conditioning composition
FI69503C (en) * 1984-03-13 1986-02-10 Neste Oy YTBELAGD BERGSBEHAOLLARE ELLER TUNNEL
US4683202A (en) * 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4965188A (en) * 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4937370A (en) * 1987-06-02 1990-06-26 The Procter & Gamble Company Novel chromophores, sunscreen compositions and methods for preventing sunburn
US4999186A (en) * 1986-06-27 1991-03-12 The Procter & Gamble Company Novel sunscreen agents, sunscreen compositions and methods for preventing sunburn
US5322770A (en) * 1989-12-22 1994-06-21 Hoffman-Laroche Inc. Reverse transcription with thermostable DNA polymerases - high temperature reverse transcription
WO1988002025A1 (en) * 1986-09-17 1988-03-24 Sri International Expression vectors for bacilli
US5087372A (en) * 1989-03-24 1992-02-11 Asahi Kasei Kogyo Kabushiki Kaisha Method for removing heavy metal ions from contaminated water and a porous membrane usable therefor
US5011681A (en) * 1989-10-11 1991-04-30 Richardson-Vicks, Inc. Facial cleansing compositions
US5073372A (en) * 1990-11-30 1991-12-17 Richardson-Vicks, Inc. Leave-on facial emulsion compositions
US5073371A (en) * 1990-11-30 1991-12-17 Richardson-Vicks, Inc. Leave-on facial emulsion compositions
US5436228A (en) * 1990-12-12 1995-07-25 Postlethwaite; Arnold E. Chemotactic wound healing peptides
GB9415421D0 (en) 1994-07-30 1994-09-21 Procter & Gamble Cosmetic compositions and processes for manufacture thereof
DK0794764T3 (en) 1994-11-28 1999-10-11 Procter & Gamble Topical skin care compositions containing thickened polyol carboxylic acid esters as skin conditioners
JP2001519765A (en) * 1996-04-18 2001-10-23 ザ ジェネラル ホスピタル コーポレイション Regulation of epithelial-mesenchymal interaction
US5827508A (en) * 1996-09-27 1998-10-27 The Procter & Gamble Company Stable photoprotective compositions
US5976838A (en) * 1996-12-18 1999-11-02 Genetics Institute, Inc. Secreted proteins and polynucleotides encoding them
CZ181199A3 (en) 1996-11-22 1999-10-13 The Procter & Gamble Company Cosmetic preparation being suitable for local application to skin or hair
DE19735587B4 (en) * 1997-08-16 2012-03-22 Eberhard-Karls-Universität Tübingen Universitätsklinikum A peptide having a radioprotective effect, a cosmetic or pharmaceutical composition containing the same, a nucleic acid encoding the same, a preparation process for this peptide and the use as a radioprotective agent
US6747137B1 (en) * 1998-02-13 2004-06-08 Genome Therapeutics Corporation Nucleic acid sequences relating to Candida albicans for diagnostics and therapeutics
HUP0102457A2 (en) * 1998-07-20 2001-10-28 M & E Biotech A/S Novel methods for the identification of ligand and target biomolecules
US5989528A (en) 1998-07-30 1999-11-23 The Procter & Gamble Company Sunscreen compositions
US5935556A (en) * 1998-07-30 1999-08-10 The Procter & Gamble Company Sunscreen compositions
JP3855042B2 (en) * 1998-09-22 2006-12-06 独立行政法人産業技術総合研究所 Hair identification method
US5968485A (en) * 1998-10-16 1999-10-19 The Procter & Gamble Company UV protection compositions
US5972316A (en) * 1998-10-16 1999-10-26 The Procter & Gamble Company UV protection compositions
WO2000024371A1 (en) 1998-10-23 2000-05-04 The Procter & Gamble Company Skin care compositions
GB9825854D0 (en) * 1998-11-25 1999-01-20 Univ Bristol Peptide
JP3106191B1 (en) * 1999-03-30 2000-11-06 工業技術院長 Physiological function controlling peptide of FGF-5 and pharmaceutical composition containing the peptide
US7090973B1 (en) * 1999-04-09 2006-08-15 Oscient Pharmaceuticals Corporation Nucleic acid sequences relating to Bacteroides fragilis for diagnostics and therapeutics
US20040031072A1 (en) * 1999-05-06 2004-02-12 La Rosa Thomas J. Soy nucleic acid molecules and other molecules associated with transcription plants and uses thereof for plant improvement
US20100293669A2 (en) * 1999-05-06 2010-11-18 Jingdong Liu Nucleic Acid Molecules and Other Molecules Associated with Plants and Uses Thereof for Plant Improvement
AU5056501A (en) * 2000-03-31 2001-10-08 Nobuyuki Itoh Fibroblast growth factor-like molecules and uses thereof
AU2001257012A1 (en) * 2000-04-14 2001-10-30 Genencor International, Inc. Methods for selective targeting
US20110131679A2 (en) * 2000-04-19 2011-06-02 Thomas La Rosa Rice Nucleic Acid Molecules and Other Molecules Associated with Plants and Uses Thereof for Plant Improvement
US6822082B2 (en) * 2000-06-30 2004-11-23 Zymogenetics, Inc. Polynucleotides for mammalian secreted protein, Z1055G2P
JP4493271B2 (en) * 2000-12-13 2010-06-30 アナフォア インコーポレイテッド Combinatorial library of proteins having a skeleton structure of C-type lectin-like domain
US7214786B2 (en) * 2000-12-14 2007-05-08 Kovalic David K Nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
CA2434323A1 (en) * 2001-02-07 2002-11-28 Centre National De La Recherche Scientifique (Cnrs) Sequence of the photorhabdus luminescens strain tt01 genome and uses
CA2515599A1 (en) * 2003-02-11 2004-08-26 Institut Pasteur Identification and characterization of racemases, definition of protein signatures, and a test for detecting d-amino acid and for screening molecules capable of inhibiting the activity of racemase, especially proline racemase
EP1680507B1 (en) * 2003-11-06 2011-06-22 Genencor International, Inc. Bacterial expression of protease inhibitors and variants thereof
ATE555119T1 (en) * 2003-11-06 2012-05-15 Danisco Us Inc EXPRESSION OF PROTEASE INHIBITORS AND VARIANTS THEREOF IN FILAMENTOUS FUNGI
US7319142B1 (en) * 2004-08-31 2008-01-15 Monsanto Technology Llc Nucleotide and amino acid sequences from Xenorhabdus and uses thereof
US7772181B2 (en) * 2008-10-15 2010-08-10 Danisco Us Inc. Personal care compositions comprising modified variant Bowman Birk Protease Inhibitors
US7803902B2 (en) * 2008-10-15 2010-09-28 Danisco Us Inc. Modified variant bowman birk protease inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5411873A (en) * 1984-05-29 1995-05-02 Genencor, Inc. Process for producing heterologous polypeptides
US5679543A (en) * 1985-08-29 1997-10-21 Genencor International, Inc. DNA sequences, vectors and fusion polypeptides to increase secretion of desired polypeptides from filamentous fungi
US5429950A (en) * 1992-03-30 1995-07-04 Genencor International, Inc. Heterologous gene expression in bacillus subtilis: fusion approach
US6063611A (en) * 1996-03-12 2000-05-16 Genencor International, Inc. Alkaline cellulase and method of producing same
US6872563B1 (en) * 1999-10-05 2005-03-29 President And Fellows Of Harvard College Compositions and methods for production of disulfide bond containing proteins in host cells
US6537968B1 (en) * 2000-07-24 2003-03-25 Alphamed Pharmaceuticals Corp Treatment of lupus erythematosus

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10087451B2 (en) 2006-09-22 2018-10-02 Aviex Technologies Llc Live bacterial vectors for prophylaxis or treatment
US11485773B1 (en) 2009-02-09 2022-11-01 David Gordon Bermudes Protease inhibitor:protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US9657085B1 (en) 2009-02-09 2017-05-23 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US10590185B1 (en) 2009-02-09 2020-03-17 David Gordon Bermudes Protease inhibitor: protease sensitive expression system and method improving the therapeutic activity and specificity of proteins and phage and phagemids delivered by bacteria
US10954521B1 (en) 2010-02-09 2021-03-23 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US10364435B1 (en) 2010-02-09 2019-07-30 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US9878023B1 (en) 2010-02-09 2018-01-30 David Gordon Bermudes Protease inhibitor: protease sensitive expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
US10857233B1 (en) 2010-02-09 2020-12-08 David Gordon Bermudes Protease inhibitor combination with therapeutic proteins including antibodies
US9486513B1 (en) 2010-02-09 2016-11-08 David Gordon Bermudes Immunization and/or treatment of parasites and infectious agents by live bacteria
US11219671B1 (en) 2010-02-09 2022-01-11 David Gordon Bermudes Protease inhibitor:protease sensitive expression system, composition and methods for improving the therapeutic activity and specificity of proteins delivered by bacteria
US10501746B1 (en) 2013-02-14 2019-12-10 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US9593339B1 (en) 2013-02-14 2017-03-14 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US11827890B1 (en) 2013-02-14 2023-11-28 David Gordon Bermudes Bacteria carrying bacteriophage and protease inhibitors for the treatment of disorders and methods of treatment
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria

Also Published As

Publication number Publication date
US7524816B2 (en) 2009-04-28
ATE530560T1 (en) 2011-11-15
ATE513917T1 (en) 2011-07-15
WO2005047314A2 (en) 2005-05-26
DE602004029812D1 (en) 2010-12-09
US20110230405A1 (en) 2011-09-22
US20130065829A1 (en) 2013-03-14
ES2375360T3 (en) 2012-02-29
ATE486087T1 (en) 2010-11-15
US7413877B2 (en) 2008-08-19
WO2005046709A2 (en) 2005-05-26
US7485618B2 (en) 2009-02-03
WO2005047314A3 (en) 2006-04-20
JP2008500805A (en) 2008-01-17
JP5728329B2 (en) 2015-06-03
EP1692158B1 (en) 2011-10-26
JP4843497B2 (en) 2011-12-21
US20050203026A1 (en) 2005-09-15
WO2005046710A2 (en) 2005-05-26
EP1692160A2 (en) 2006-08-23
EP1692159B1 (en) 2010-09-29
US20050238607A1 (en) 2005-10-27
ATE482972T1 (en) 2010-10-15
CA2544820C (en) 2014-02-18
DK1692158T3 (en) 2012-02-06
EP1680507B1 (en) 2011-06-22
DE602004029393D1 (en) 2010-11-11
US20050202535A1 (en) 2005-09-15
US7696173B2 (en) 2010-04-13
WO2005047511A2 (en) 2005-05-26
EP1692160B1 (en) 2010-10-27
US20080113917A1 (en) 2008-05-15
JP2012024091A (en) 2012-02-09
WO2005047511A3 (en) 2005-11-24
EP1680507A2 (en) 2006-07-19
DK1680507T3 (en) 2011-10-10
WO2005046710A3 (en) 2005-11-24
EP1692158A2 (en) 2006-08-23
CA2544820A1 (en) 2005-05-26
EP1692159A2 (en) 2006-08-23
US20080269139A1 (en) 2008-10-30
WO2005046709A3 (en) 2005-11-24

Similar Documents

Publication Publication Date Title
US7696173B2 (en) Compositions comprising bowman-birk protease inhibitors and variants thereof
JP2008500805A5 (en)
JP7134629B2 (en) Proteases with modified propeptide regions
Matsui et al. Leucine aminopeptidases: diversity in structure and function
EP2344531B1 (en) Modified variant bowman birk protease inhibitors
Pyati et al. Protein digestion in cereal aphids (Sitobion avenae) as a target for plant defence by endogenous proteinase inhibitors
RU2515112C2 (en) Improved production of protein in bacillus
US20110171718A1 (en) Proteases With Modified Pre-Pro Regions
JP2916228B2 (en) Method of preparing microorganisms of genetically engineered recombinant aprotinin variants, homogeneously processed aprotinin variants and their therapeutic use
Lütticke et al. E. coli LoiP (YggG), a metalloprotease hydrolyzing Phe–Phe bonds
Malik et al. Periplasmic production of native human proinsulin as a fusion to E. coli ecotin
Vogtentanz et al. A Bacillus subtilis fusion protein system to produce soybean Bowman–Birk protease inhibitor
García-Fernández et al. Two variants of the major serine protease inhibitor from the sea anemone Stichodactyla helianthus, expressed in Pichia pastoris
MacKenzie et al. Aberrant processing of wild-type and mutant bovine pancreatic trypsin inhibitor secreted by Aspergillus niger
Jankiewicz et al. Biochemical characterization of an alkaline metallopeptidase secreted by a Pseudomonas fluorescens isolated from soil
Kojima et al. Involvement of the C-terminal region of yeast proteinase B inhibitor 2 in its inhibitory action
Inoue et al. Heterologous expression and site‐directed mutagenesis studies on the activation mechanism and the roles of the basic residues in the prosegment of aspergillopepsinogen I
KR930003913B1 (en) Method for preparation of polypeptide having thrombin inhibiting activity
Tanaka et al. High-level production and purification of clostripain expressed in a virulence-attenuated strain of Clostridium perfringens
JPH0648988B2 (en) Method for producing thrombin inhibitor
CN114686519A (en) Method for preparing serine protease
JPWO2006064721A1 (en) Low temperature activated subtilisin
Whitaker Enzymes in Protein Biosynthesis
Na et al. Purification and characterization of an extracellular protease from culture filtrate of salmonella schttmulleri
Kresze I. PROTEOLYSIS OF NATIVE AND DENATURED PROTEINS

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENCOR INTERNATIONAL, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COLLIER, KATHERINE;GANSHAW, GRANT;DE NOBEL, HANS;AND OTHERS;REEL/FRAME:021404/0581;SIGNING DATES FROM 20050404 TO 20050519

Owner name: GENENCOR INTERNATIONAL, INC.,CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:COLLIER, KATHERINE;GANSHAW, GRANT;DE NOBEL, HANS;AND OTHERS;SIGNING DATES FROM 20050404 TO 20050519;REEL/FRAME:021404/0581

AS Assignment

Owner name: DANISCO US INC.,CALIFORNIA

Free format text: CHANGE OF NAME;ASSIGNOR:GENENCOR INTERNATIONAL, INC.;REEL/FRAME:023893/0996

Effective date: 20070215

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552)

Year of fee payment: 8

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 12TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1553); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 12